US20080124760A1 - Regulatory Nucleic Acid Elements - Google Patents

Regulatory Nucleic Acid Elements Download PDF

Info

Publication number
US20080124760A1
US20080124760A1 US11/762,240 US76224007A US2008124760A1 US 20080124760 A1 US20080124760 A1 US 20080124760A1 US 76224007 A US76224007 A US 76224007A US 2008124760 A1 US2008124760 A1 US 2008124760A1
Authority
US
United States
Prior art keywords
seq
nucleic acid
gene
expression
sequence
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/762,240
Other languages
English (en)
Inventor
Barbara Enenkel
Kerstin Sautter
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Boehringer Ingelheim Pharma GmbH and Co KG
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Publication of US20080124760A1 publication Critical patent/US20080124760A1/en
Priority to US12/842,468 priority Critical patent/US9045776B2/en
Assigned to BOEHRINGER INGELHEIM PHARMA GMBH & CO. KG reassignment BOEHRINGER INGELHEIM PHARMA GMBH & CO. KG ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SAUTTER, KERSTIN, ENENKEL, BARBARA
Priority to US14/699,182 priority patent/US9708626B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/79Vectors or expression systems specially adapted for eukaryotic hosts
    • C12N15/85Vectors or expression systems specially adapted for eukaryotic hosts for animal cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • C12N15/67General methods for enhancing the expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2800/00Nucleic acids vectors
    • C12N2800/10Plasmid DNA
    • C12N2800/106Plasmid DNA for vertebrates
    • C12N2800/107Plasmid DNA for vertebrates for mammalian
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/46Vector systems having a special element relevant for transcription elements influencing chromatin structure, e.g. scaffold/matrix attachment region, methylation free island
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2830/00Vector systems having a special element relevant for transcription
    • C12N2830/80Vector systems having a special element relevant for transcription from vertebrates
    • C12N2830/85Vector systems having a special element relevant for transcription from vertebrates mammalian

Definitions

  • the invention relates to cis-active nucleic acid sequences, so-called TE elements.
  • the TE elements preferably originate from the CHO genome. Their use in expression vectors, for example, in stable cell populations permits at least twice as high an expression of a gene of interest in a desired chromosome locus compared with vectors previously used.
  • Mammalian cells are the preferred host cells for the production of complex biopharmaceutical proteins as the post-translational modifications are human-compatible both functionally and from a pharmacokinetic point of view.
  • the main relevant cells types are hydridomas, myelomas, CHO (Chinese Hamster Ovary) cells and BHK (Baby Hampster Kidney) cells.
  • the host cells are increasingly cultivated under serum- and protein-free production conditions. The reasons for this are the associated reduction in costs, the reduced interference in the purification of the recombinant protein and the reduction of the potential for introducing pathogens (e.g. prions and viruses).
  • the use of CHO cells as host cells is becoming more and more widespread as these cells adapt to suspension growth in serum- and protein-free medium and moreover are regarded and accepted as safe production cells by the regulatory bodies.
  • the heterologous gene is generally introduced into the desired cell line together with a selectable marker gene, such as neomycin phosphotransferase (NPT), by transfection.
  • a selectable marker gene such as neomycin phosphotransferase (NPT)
  • the heterologous gene and the selectable marker gene can be expressed in a host cell, starting from an individual or separate co-transfected vectors. Two to three days after the transfection the transfected cells are transferred into medium containing a selective agent, e.g. G418 when using the neomycin-phosphotransferase gene (NPT gene) and cultivated for a few weeks under these selective conditions.
  • the emergent resistant cells which have integrated the exogenous DNA can be isolated and investigated for the expression of the desired gene product (gene of interest).
  • the expression vectors for production cells are equipped with strong, usually constitutively expressing promoters and enhancers such as CMV enhancer and promoter, for example, to allow high product expression.
  • promoters and enhancers such as CMV enhancer and promoter, for example, to allow high product expression.
  • CMV enhancer and promoter for example, to allow high product expression.
  • cells are selected which have the product gene stably integrated in their genome. This is done with selectable markers such as e.g. neomycin-phosphotransferase (NPT) and dihydrofolate reductase (DHFR).
  • NPT neomycin-phosphotransferase
  • DHFR dihydrofolate reductase
  • LCR locus control regions
  • LCRs are capable of opening chromatin in their native tissue (Ortiz et al., 1997).
  • ⁇ -thalassaemia in which the ⁇ -globin locus is intact but is not expressed.
  • the reason for the lack of expression is a major deletion in the 5′ direction of the ⁇ -globin genes.
  • the deletion of this ⁇ -globin LCR leads to a closed chromatin configuration which extends over the entire locus and leads to suppression of gene expression (Li et al., 2002).
  • LCRs colocalise with DNAse I-hypersensitive sites (HS) in the chromatin of expressing cells. The occurrence of HS also indicates open chomatin.
  • the HS contain a series of different general and tissue-specific binding sites for transcription factors. By the interaction of the transcription factors with the DNA the open chromatin structure of HS is produced (Li et al., 2002).
  • Many LCRs are known to be made up of a number of HS the functions of which can be more or less separated from one another.
  • the TCR ⁇ gene for example is expressed under endogenous control only in T-cell tissue.
  • the locus exists in various chomatin modes depending on the tissue and expression status. In the 3′ region it has a locus control region which has 8 HS.
  • HS 2-6 a 6 kb partial fragment of the LCR, has a chromatin-opening activity and is not tissue specific.
  • the tissue specificity is imparted to the T-cell-specific expression in the thymus by HS7, 8 and 1 (3 kb). Only in the complete combination of all the HS is the TCR ⁇ LCR functionally complete (Ortiz et al., 1997). A more precise subdivision and specification of the individual HS functions of the TCR ⁇ LCR can be found in (Ortiz et al., 1999).
  • LCRs are functionally very complex and may be made up of different control elements such as enhancers, silencers and isolators.
  • Other Examples of the division of the LCR functions between various domains are the TCRy locus and ⁇ -globin locus.
  • the former is made up of the DNAse I-hypersensitive site HsA and the enhancer 3′E C ⁇ 1 .
  • the TCR ⁇ -LCR in addition to having its usual functions, is also thought to play a part in the recombination of the TCR ⁇ genes (Baker et al., 1999).
  • the ⁇ -globin locus has five HS with distinguishable functions which also require the tissue-specific promoter in order to function fully.
  • LCRs could also play another important role in the tissue-specific demethylation of DNA, as DNA methylation results in a closed chromatin structure and the inactivation of genes. A mechanism of activity which activates gene expression by increased histone acetylation would also be possible (Li et al., 2002).
  • Scaffold/Matrix Attachment Regions are DNA sequences which bind with a high affinity in vitro to components of the matrix or the scaffold of the cell nucleus. They form the structural and possibly also functional boundaries of chromatin domains (Zahn-Zabal et al., 2001). S/MARs are capable of interacting with enhancers and of locally increasing the accessibility of the DNA in the chromatin and in this way can increase the expression of stably integrated heterologous genes in cell lines, transgenic animals and plants (Klehr et al., 1991; Stief et al., 1989; Jenuwein et al., 1997; Zahn-Zabal et al., 2001).
  • MARs have also been reported which do not impart high expression but play an important part in the correct regulation of development-specific genes (McKnight et al., 1992).
  • Isolators are defined as a neutral boundary between neighbouring regions which influence one another, e.g. between active and inactive chromatin (boundary elements). They may restrict the effect of enhancers or isolate entire DNA domains against them and shield stably transfected reporter genes from positional effects (Bell and Felsenfeld, 1999; Udvardy, 1999). Thus, these elements render the expression independent of the genomic position. They may also prevent the silencing of transgenes in the absence of selection pressure (Pikaart et al., 1998). Another presumed function of isolators is the restriction of replication territories (Bell and Felsenfeld, 1999). The first isolators that were described are scs and scs' from Drosophila. They constitute the boundary for the hsp70 heat shock genes and suppress positional effects (Udvardy et al., 1985).
  • a GC-rich fragment from the dhfr gene (Chinese Hamster) was found, containing CpG islands (Poljak et al., 1994). The fragment on its own exhibited no influence whatever on reporter gene expression. Situated between an expression promoting SAR and the reporter gene, however, the fragment was able to substantially prevent the expression-enhancing effect of the SAR element. Possibly, this GC-rich fragment blocks the chromatin-opening mechanism of the SAR element and consequently acts as an isolator. Elements with extended CPG islands are methylated with a higher probability as they are recognised by a DNA methyltransferase which converts cytosine into 5-methylcytosine. Consequently, inactive chomatin is formed (Poljak et al., 1994).
  • Aronow and colleagues defined in the first intron of the human ADA gene (Adenosin deaminase) a new regulatory element which substantially contributes to expression which is dependent on gene copy number but independent of position (Aronow et al., 1995).
  • the element is up to 1 kb in size and only functional when it flanks a 200 bp T-cell-specific enhancer. If only one of the two segments is present or if the segments are wrongly arranged in their sequence and orientation, the element is non-functional as this prevents the formation of DNAse I-hypersensitive sites on the enhancer.
  • the Ubiquitous Chromatin Opening Elements are responsible for an open chromatin structure in chromosomal regions with ubiquitously expressed household genes (human hnRNP A2 gene, human ⁇ -actin gene, human PDCD2 gene). All these genes have CpG-rich islands in the untranslated regions which are relatively weakly methylated. The absence of methylation of CpG islands indicates that there is active chromatin at this point.
  • the UCOEs help to provide a strength of expression which is independent of the genomic environment and the nature of the cell or tissue.
  • the firm Immunex also describes cis-active DNA sequences which bring about an increase in expression (U.S. Pat. No. 6,027,915, U.S. Pat. No. 6,309,851).
  • the element referred to as the expression augmenting sequence element (EASE) demands a high expression of recombinant proteins in mammalian cells, is not active in transient expression systems and does not have the typical sequence properties found in LCRs and S/MARs. It is also not a sequence which codes for a trans-activating protein as it does not contain an open reading frame.
  • the fragment is 14.5 kb long, originates from the genomic DNA of CHO cells and can increase the expression of a stably integrated reporter gene eight fold.
  • HMG-I(Y) proteins belong to the family of the high mobility group non-histone chromatin proteins. They are also referred to as “architechtonic transcription factors” and form a new category of trans-regulators of mammalian genes. HMG-I(Y) proteins recognise 80-rich sequences and bind to their so-called AT hooks (DNA-binding domains) in the small DNA fork. This can lead to local changes in the DNA topology and consequently to altered gene expression.
  • STAR elements Stimulatory and Anti-Repressor Elements
  • the screening was carried out using a specially designed reporter plasmid.
  • the expression of the reporter gene was only possible when it was functionally linked with an anti-repressor element from the human gene bank.
  • the STAR elements thus obtained the authors were able to protect transgenes from positional effects in the genome of mammalian cells.
  • a comparison with the mouse genome showed that the majority of these STAR elements occur in both the human and the murine genome and are highly conserved within these two species.
  • a major problem in establishing cell lines with a high expression of the desired protein arises from the random and undirected integration of the recombinant vector in transcription-active or -inactive loci of the host cell genome. As a result, a population of cells is obtained which show completely different expression rates for the heterologous gene, while the productivity of the cells generally follows a normal distribution.
  • In order to identify cell clones which have a very high expression of the heterologous gene of interest it is therefore necessary to check and test a number of clones, resulting in high expenditure of time, labour and costs. Attempts at improving the vector system used for the transfection are therefore directed at even allowing or increasing the transcription of a stably integrated transgene by the use of suitably cis-active elements.
  • the cis-active elements which act at the chromatin level include for example the locus control regions, scaffold-matrix attachment regions, isolators, etc., already described. Some of these elements shield certain genes from the influences of the surrounding chromatin. Others exhibit an enhancer-like activity, although this is restricted to stably integrated constructs.
  • Stable cell lines with high productivity are usually produced by selection with positive selectable markers, frequently combined with agent-induced gene amplification (e.g. dihydrofolate reductase/methotrexate or glutamine synthetase/methioninesulfoximine).
  • agent-induced gene amplification e.g. dihydrofolate reductase/methotrexate or glutamine synthetase/methioninesulfoximine.
  • the pools and clones which are produced with this selection strategy are investigated for high and stable expression in a complex screening process.
  • the majority of the clones produce no or only average amounts of product and only a few are high producers.
  • the proportion of high producers in a mixed population can be increased, for example, by a mutation in the selectable marker (Sautter und Enenkel, 2005, WO 2004/050884).
  • the present invention relates to regulatory nucleic acids, particularly a nucleic acid having SEQ ID No. 1, known as a “TE element”, or a fragment or derivative thereof, which leads to an increase in the transcription or expression of a gene of interest in stably transfected cells.
  • a TE element of this kind on an expression vector in conjunction with a promoter, a product gene, a selectable marker and optionally an enhancer in stable integration into a host genome, such as the CHO-DG44 genome, for example, overcomes, shields or cancels out the chromosomal positional effects.
  • both the proportion of high producers in a transfection batch and also the absolute expression level are increased.
  • the invention further relates to expression vectors which contain transcription- or expression-increasing regions, fragments or derivatives of SEQ ID No. 1, preferably the TE elements TE-00 (SEQ ID No. 2), TE-01 (SEQ ID No. 3), TE-02 (SEQ ID No. 4), TE-03 (SEQ ID No. 5), TE-04 (SEQ ID No. 6), TE-06 (SEQ ID No. 8), TE-07 (SEQ ID No. 9), TE-08 (SEQ ID No. 10), TE-10 (SEQ ID No. 12), TE-11 (SEQ ID No. 13), TE-12 (SEQ ID No. 14).
  • TE-06 TE-07 and TE-08 re particularly preferred.
  • SEQ ID No. 1 originates from a sequence region located upstream of the coding region of the ubiquitin/S27agene, which was isolated from CHO— cells, the gene coding for an essential protein in the ribosome metabolism of the cell.
  • cis-active TE elements Compared with the expression vectors used hitherto, the additional introduction of the cis-active TE elements into expression vectors results in a productivity of stably transfected cell pools, particularly CHO-DG44 cell pools, which is up to seven times higher.
  • transient transfections of CHO-DG44 cell pools no increase in productivity can be achieved by introducing the TE elements.
  • the increase in productivity observed in the stable cell pools is not based on an enhancer present in the TE elements.
  • chromosomal integration is absolutely essential for the increase in productivity caused by TE elements. This is an indication that TE elements may suppress, shield or cancel out negative chromosomal positional effects.
  • cis-active elements have been produced and identified which are characterised by their particular suitability for selecting and enriching high producing cells and are therefore capable of reducing the expenditure on time, costs and capacity in the isolation and identification of high producing clones.
  • Possible applications for the invention include the development of high producing cell lines as required for example in the manufacture of biopharmaceuticals, in analytical cell-based assays, in high throughput screenings of substances or in the production of recombinant protein products for NMR spectroscopy, other assays, etc. Because of the higher specific productivity and the reduction of cells which express little or no product, more and higher producing cell lines can be established in a shorter time and thus labour and costs can be saved. Other possible application are the production of robust improved host cell lines (e.g. the introduction of anti-apoptosis or glycosilation genes), transgenic animals or plants and in gene therapy.
  • robust improved host cell lines e.g. the introduction of anti-apoptosis or glycosilation genes
  • the invention does not arise from the prior art.
  • the nucleic acid with SEQ ID No. 1 is a nucleic acid sequence isolated from the genome of Chinese hamsters ( Cricetulus griseus ). It comes from a sequence region located upstream of the coding region of the ubiquitin/S27agene.
  • the nucleic acid with SEQ ID No. 1 has an average GC content of 44% and does not contain any lengthy passages of GC repeats. This GC content is comparable with the average GC content of about 40% described for genomic DNA of mammals (Delgado et al., 1998).
  • nucleotides 1579 to 3788 of SEQ ID No. 1 correspond to the nucleotides 1 to 2201 of SEQ ID No.5 from WO 97/15664, but with a difference.
  • SEQ ID No.1 four additional nucleotides were introduced, as a result of the cloning process, formed by a reaction of filling an existing ECORI cutting site. This insertion of the additional four nucleotides took place between nucleotide 357 and 358 of sequence SEQ ID No.5 from WO 97/15664.
  • Nucleotides 1 to 1578 of the nucleic acid sequence with SEQ ID No.1 from the present invention constitute new hitherto unknown sequence regions which were isolated within the scope of this invention.
  • WO 97/15664 did not disclose that SEQ ID No.1 from the present invention or fragments or derivatives thereof increase the transcription or expression of a gene of interest irrespective of the chromosomal integration site when they are functionally linked to a promoter/enhancer combination which allows the transcription of the functionally linked gene of interest.
  • WO 97/15664 discloses the use of 5′UTR sequences of the ubiquitin/S27a gene as promoter, while the sequence region from position ⁇ 161 to ⁇ 45 according to FIG.
  • nucleic acid of SEQ ID No.1 according to the invention when using standard alignment algorithms such as BLAST, show no sequence homologies with the nucleic acids sequences described in the following patent applications, which can also positively influence the expression at the chromatin level in cis:
  • FIG. 1 SCHEMATIC REPRESENTATION OF THE BASE VECTORS
  • E/P in this case is a combination of CMV enhancer and hamster ubiquitin/S27a promoter
  • P is merely a promoter element
  • T is a termination signal for the transcription which is necessary for the polyadenylation of the transcribed mRNA.
  • the position and direction of transcription initiation within each transcription unit is indicated by an arrow.
  • SpeI SpeI cutting site
  • the amplifiable selectable marker dihydrofolate reductase is abbreviated to “DHFR”.
  • the selectable marker neomycin phosphotransferase contains the point mutation D227G and is abbreviated to “D227G” accordingly in the Figure.
  • the “IRES” element originating from the encephalomyocarditis virus acts as an internal ribosomal binding site within the bicystronic transcription unit and allows translation of the following green fluorescent protein “GFP”.
  • GFP green fluorescent protein
  • HC and LC code for the heavy and light chains, respectively, of a humanised monoclonal IgG1 antibody.
  • E/P is a combination of CMV enhancer and CMV promoter
  • P is merely a promoter element
  • T is a termination signal for the transcription which is needed for the polyadenylation of the transcribed mRNA.
  • the position and direction of transcription initiation within each transcription unit is indicated by an arrow.
  • a sequence region “A” with cutting sites for restriction endonucleases (adapter) is inserted before the promoter.
  • the selectable marker neomycin phosphotransferase contains the point mutation F2401 and is accordingly abbreviated to F2401 in the figure.
  • the IRES element originating from the Encephalomyocarditis virus acts as an internal ribosomal binding site within the bicistronic transcription unit and allows translation of the subsequent red fluorescent protein “dsRed”.
  • MCP-1 codes for human monocyte chemoattractant Protein-1.
  • FIG. 2 SCHEMATIC REPRESENTATION OF AN MCP-1 BASE VECTOR
  • E/P is a combination of “E/P” is a combination of CMV enhancer and CMV promoter, “P” is merely a promoter element and “T” is a termination signal for the transcription which is needed for the polyadenylation of the transcribed mRNA.
  • T is a termination signal for the transcription which is needed for the polyadenylation of the transcribed mRNA.
  • the position and direction of transcription initiation within each transcription unit is indicated by an arrow.
  • a sequence region “A” with cutting sites for restriction endonucleases (adapter) is inserted before the promoter.
  • the selectable marker dihydrofolate reductase is abbreviated to “dhfr” in the figure.
  • the IRES element originating from the Encephalomyocarditis virus acts as an internal ribosomal binding site within the bicistronic transcription unit and allows translation of the subsequent red fluorescent protein “dsRed”.
  • MCP-1 codes for human monocyte chemoattractant Protein-1.
  • FIG. 3 5′ SEQUENCE OF THE CHO UBIQUITIN/S27S GENE
  • sequence region comprising 3788 bp was isolated from the genome of CHO (Chinese Hamster Ovary) cells and is located upstream of the coding region of the Ub/S27a gene, which is a fusion between a ubiquitin unit (Ub) and a ribosomal protein of the small ribosome subunit (S27a).
  • FIG. 4 GRAPHIC REPRESENTATION OF THE TE ELEMENTS 00 TO 12
  • This figure schematically shows the genomic sequence region of 3788 bp, which was subcloned in a plasmid, located upstream of the coding region of the CHO ubiquitin/S27a gene.
  • SEQ ID No. 1 also known as TE element A
  • TE elements partial fragments of different lengths, hereinafter referred to as TE elements, were prepared.
  • TE element 00 (SEQ ID No. 2) was isolated from a subclone of this sequence as a Sac II restriction fragment and cloned into the SpeJ cutting site of the target vectors pBID-HC and pBING-LC. These contained either the gene for the heavy chain (HC) or light chain of an IgGI (see FIG. 1A ).
  • expression vectors were formed in which the TE element 00 is positioned in direct and reversed orientation upstream of the promoter.
  • the TE elements 01 to 12 were produced by PCR with various pairs of primers (see FIGS. 5 and 6 ) and cloned into the base plasmid pTE4/MCP-1 ( FIG. 1B ) and pTE5/MCP-1 ( FIG. 2 ) via BamHI/BsrGI.
  • FIG. 5 TE ELEMENTS 00 TO 12
  • This Table shows the size and the starting and end positions of the TE elements 00 to 21, which were produced from the TE-A sequence (SEQ ID No. 1).
  • the primers used are additionally specified.
  • the size gradations of the elements are about 500 bp and have deletions at the 5′ or 3′ end, compared with the starting sequence TE-A (SEQ ID No. 1).
  • FIG. 6 PRIMER FOR SYNTHESISING THE TE ELEMENTS 01 TO 12
  • primers are shown in the 5′-3′ direction.
  • Primers with “for” in their name are primers in direct orientation of SEQ ID No. 1
  • primers with “rev” are those in reverse orientation.
  • Each primer consists at the 5′ end of six nucleotides followed by a BamHJ or BsrGI cutting site and a sequence of about 20 to 30 nucleotides which is 100% homologous with a sequence portion in SEQ ID No. 1.
  • the region of the primer homologous with SEQ ID No. 1 is shown in bold.
  • One for primer and one rev primer was used to amplify a sequence region of SEQ ID No. 1.
  • the resulting PCR product was cloned into the base plasmid pTE4/MCP-1 ( FIG. 1B ) or pTE5/MCP-1 ( FIG. 2 ) via BamHI and BsrGI.
  • FIG. 7 FACS MEASUREMENT OF THE TRANVECTION SERIES B
  • the Figure shows the relative increase in GFP expression in cells with the TE element 00 compared with cells without the TE element 00.
  • CHO-DG4 cells were transfected with the plasmid combinations pBING-LC and pBID-HC, which differ from one another only in the presence and orientation of the TE element 00.
  • the GFP fluorescence was measured by FACS analysis.
  • Each graph with the exception of the untransfected CHO-DG44 cells (DG44) serving as a negative control, constitutes the average of the GFP fluorescence from, in each case, 10 pools of transfection series B. 20000 cells were studied per pool.
  • Control denotes the base plasmids pBING-LC and pBID-HC
  • reverse denotes a reverse orientation of the TE element 00 in the base vectors
  • direct indicates a direct orientation of TE element 00 in the base vectors.
  • FIG. 8 FACS MEASUREMENT OF TRANSVECTION SERIES C
  • the Figure shows the proportion of dsRed2-expressing cells in stable cell populations which contained the TE elements 01, 02, 05, 06, 08 or 09, compared with cells in cell populations which did not contain a TE element.
  • CHO-DG44 cells were transfected with the plasmid pTE4/MCP-1 or derivatives obtained therefrom, which additionally contained one of the TE elements mentioned above. After an approximately three-week long selection of the transfected cell pools in medium with added G418, the dsRed2 fluorescence was measured by FACS analysis. 10000 cells were measured per pool and the inherent fluorescence of the untransfected CHO-DG44 cells was substracted. Each value is the average of the percentage proportion of dsRed2-expressing cells from 6 pools of transfection series C.
  • FIG. 9 INFLUENCE OF THE TE ELEMENTS ON THE SPECIFIC PRODUCTIVITY
  • This Figure shows the changes in the expression level of IgG1 or MCP-1 which are obtained as a result of the presence of the TE elements compared with control pools with no TE element, shown graphically (A) or in table form (B).
  • the cell pools were produced by stable transfection of CHO-DG44 cells with the base plasmids pBING-LC and pBID-HC or pTE4/MCP-1 (“control”) and the derivatives obtained therefrom, each of which additionally contained a TE element (“00” in direct orientation (“00 direct”) and in reverse orientation (“00 reverse”), “01” to “12”).
  • FIG. 10 INFLUENCE OF THE TE ELEMENTS ON THE SPECIFIC PRODUCTIVITY IN DHFR-SELECTED CELL POOLS
  • This Figure shows the changes in the expression levels of MCP-1 which resulted from the presence of the TE elements compared with control pools with no TE element, in the form of a graph (A) or table (B).
  • the cell pools were produced by stable transfection of CHO-DG44 cells with the base plasmid pTE5/MCP-1 (“control”) or derivatives obtained therefrom, each of which additionally contained a TE element (“01” to “12”) (Series E). After a two to three-week long selection of the transfected cell pools in HT-free medium the protein expression was measured by ELISA in the cell culture supernatant and the specific productivity was calculated per cell and per day.
  • the cultivation of the stably transfected CHO-DG44 cells was carried out by several passages in 75 cm 2 T flasks with a passaging rhythm of 2-2-3 days. Six pools of each plasmid variant through 6 passages were in cultivation. The specific productivities of the pools of a plasmid variant were averaged and the average of the controls was set at 1. The averaged specific productivities of the pools with a TE element was compared with this.
  • FIG. 11 TESTING THE TE ELEMENTS FOR ENHANCER ACTIVITY
  • TE elements 01 to 12 thus do not act as enhancers and can therefore only bring about a significant increase in expression when integrated in the chromosomes.
  • the cell culture supernatant was removed and the MCP-1 titre was determined by ELISA and the SEAP activity was determined.
  • the MCP-1 titre was corrected with regard to the transfection efficiency, determined by SEAP expression.
  • the Figure shows the average of the 6 parallel pools with standard deviation.
  • FIG. 12 OTHER TE ELEMENTS
  • FIG. 13 TESTING OF DIFFERENT POSITIONS AND COMBINATIONS OF THE TE ELEMENTS
  • This Figure represents a selection of possible expression vectors in which different positions, orientation and combinations of TE elements are used to investigate whether an additional increase in expression can be achieved in this way.
  • TE elements 06 and 08 As well as the flanking of the product gene by TE elements, a number of identical or different short TE elements are also connected up one behind the other, such as for example TE elements 06 and 08 or the new TE elements 13 and 14.
  • FIG. 14 INFLUENCE OF TE ELEMENTS TE 13 TO TE 18 ON THE SPECIFIC MCP-1 EXPRESSION
  • This Figure graphically shows the changes in the expression levels of MCP-1 which result from the presence of the TE elements compared with control pools with no TE element.
  • the cell pools are produced by stable transfection of CHO-DG44 cells with the base plasmid pTE4/MCP-1 (“control”) or derivatives obtained therefrom which additional each contained a TE element (“13” to “18”) (Series F). After a two to three-week long selection of the transfected cell pools in HT-supplemented medium +G418 (400 ⁇ g/ml) the protein expression was measured by ELISA in the cell culture supernatant and the specific productivity per cell and per day was calculated.
  • Cultivation of the stably transfected CHO-DG44 cells was carried out by several passages in 75 cm 2 T flasks with a passaging rhythm of 2-2-3 days. Of each plasmid variant, 4 pools were in cultivation over 5 to 6 passages. The specific productivities of the pools of a plasmid variant were averaged and the average of the controls was set at 1. The averaged specific productivities of the pools with a TE element were compared with this.
  • FIG. 15 INFLUENCE OF THE TE ELEMENTS AT VARIOUS POSITIONS AND IN VARIOUS COMBINATIONS ON THE EXPRESSION OF MCP-1
  • This Figure graphically shows the changes in the expression levels of MCP-1 which result from the presence and combination of different TE elements compared with control pools without a TE element.
  • the cell pools were produced by stable transfection CHO-DG44 cells with the base plasmid pTE4/MCP-1 (“control”) or derivatives obtained therefrom which additionally each contained one or two TE elements (“06 and 08, 08rev, 09rev, A”) (Series G). After two to three-week long selection of the transfected cell pools in HT-supplemented medium +G418 (300 ⁇ g/ml) the protein expression was measured by ELISA in the cell culture supernatant and the specific productivity per cell and per day was calculated.
  • the cultivation of the stably transfected CHO-DG44 cells was carried out by several passages in 6-well plates (MAT6) with a passaging rhythm of 2-2-3 days. Of each plasmid variant, 6 pools were in cultivation over 6 passages. The specific productivities of the pools of a plasmid variant were averaged and the average value of the controls was set at 1. The averaged specific productivities of the pools with a TE element were compared with this.
  • FIG. 16 TESTING OF THE TE ELEMENT TE-08 WITH IGG-4 ANTIBODIES
  • the vectors shown here were used for the expression of recombinant monoclonal IgG4 antibodies in CHO-DG44 cells.
  • E/P in this case is a combination of CMV enhancer and promoter, P is merely a promoter element and T is a termination signal for the transcription, which is required for the polyadenylation of the transcribed mRNA.
  • the position and direction of the transcription initiation within each transcription unit is indicated by an arrow.
  • the genes for the light chain (LC) and heavy chain (HC) were cloned in, in exchange for the MCP-1—IRES—dsRed2 cassette ( FIGS. 1B and 2 ).
  • the amplifiable selectable marker dihydrofolate reductase is abbreviated to “dhfr”.
  • the selectable marker neomycin-phosphotransferase contains the point mutation F2401 and is abbreviated accordingly to F2401 in the Figure.
  • TE element denotes regulatory nucleic acids.
  • TE element or “expression-enhancing element” or “transcription enhancing element” or “expression or transcription enhancing nucleic acid element” are used synonymously in the test. These terms all refer to regulatory nucleic acid sequences.
  • TE element or “expression enhancing element” or “transcription enhancing element” or “expression or transcription enhancing nucleic acid element” is meant in particular Sequence ID No. 1, including the complementary sequence thereto, which was isolated from the genome of the Chinese hamster ( Cricetulus griseus ), or any part, fragment or region thereof or a derivative of Sequence ID No. 1 or one of the parts, fragments or regions thereof, which when stably integrated in the chromosomes leads to an increase in the transcription or expression of a gene of interest. Also meant are any desired combinations of parts, fragments, regions or derivatives of Sequence ID No. 1 which consist of a number of identical or different parts, fragments, regions or derivatives of SEQ ID No.
  • TE element may refer both to SEQ ID No. 1 itself and to any desired fragments, parts, regions or derivatives thereof.
  • TE element transcription enhancing or expression enhancing nucleic acid element or fragments, parts, regions or derivatives thereof encompasses, in addition to parts of the sequence of the Chinese hamster ( Cricetulus griseus ), corresponding functional homologous nucleotide sequences from other organisms. Examples of these other organisms include man, mouse, rat, monkey and other mammals and rodents, reptiles, birds, fishes and plants.
  • fragment or “part” or “region” (these terms being used synonymously) is meant a nucleic acid molecule (single or double stranded) which is 100% identical in its sequence to a part of SEQ ID No. 1 or the complementary sequence thereto. It is known that the cloning of fragments which are produced either by digestion with restriction enzymes or by PCR can lead to modifications in the end regions of the fragment, i.e. additional or absent nucleotides or nucleotides additionally introduced through primers, which are the result of filling or breakdown reactions. These variations in the end regions of the fragments are included in the definition of a fragment, even if these sequence regions have a sequence identity of less than 100% with SEQ ID No. 1.
  • “Parts” or “fragments” or “regions” of Sequence ID No. 1 include for example TE-00 (Sequence ID No. 2), TE-01 (Sequence ID No. 3), TE-02 (Sequence ID No. 4), TE-03 (Sequence ID No. 5), TE-04 (Sequence ID No. 6), TE-05 (Sequence ID No. 7), TE-06 (Sequence ID No. 8), TE-07 (Sequence ID No. 9), TE-08 (Sequence ID No. 10), TE-09 (Sequence ID No. 11), TE-10 (Sequence ID No. 12), TE-11 (Sequence ID No.
  • the fragment leads to an increase in the transcription or expression of a functionally linked gene of interest.
  • “Parts” or “fragments” or “regions” of Sequence ID No. 1, which lead to an increase in the transcription or expression of a gene of interest, are for example TE-00 (Sequence ID No.
  • TE-00 in any desired orientation which lead to an increase in the transcription or expression of a gene of interest, particularly those which are wholly or at least partially in the 5′ region of TE-00 (SEQ ID No. 2). This corresponds to the partial region of SEQ ID No. 1 between 1 bp and 1578 bp. Also preferred is the fragment TE-08 (SEQ ID No. 10).
  • a “derivative” is meant, in the present invention, a nucleic acid molecule (single or double stranded) which has at least 70% sequence identity, preferably at least about 80% sequence identity, particularly preferably at least about 90% sequence identity and most preferably at least about 95% sequence identity with SEQ ID No. 1 or the complementary sequence thereto or with a part or fragment or region of SEQ ID No. 1 or the complementary sequence thereto, and which, on chromosomal integration, leads to an increase in the transcription or expression of a gene of interest. Sequence differences from SEQ ID No. 1 may be based on the one hand on differences in homologous endogenous nucleotide sequences from other organisms.
  • nucleotide acid sequence may also be based on deliberate modifications of the nucleotide acid sequence, e.g. on substitution, insertion or deletion of at least one or more nucleotides.
  • Deletion, insertion and substitution mutants can be produced by “site-specific mutagenesis” and/or “PCR-based mutagenesis techniques”. Corresponding methods are described by way of example by Lottspeich and Zorbas (1998; Chapter 36.1 with further references).
  • sequence identity can be brought into conformity with a reference sequence, in this case Sequence ID No. 1, using so-called standard alignment algorithms such as for example “BLAST” (Altschul, S. F., Gish, W., Miller, W., Myers, E. W. & Lipman, D.J.
  • a “derivative” is meant, according to the present invention, a nucleic acid molecule (single or double stranded) which hybridises with SEQ ID No. 1 or with the sequence of a fragment or part or region of SEQ ID No. 1 or of a sequence complementary thereto.
  • the hybridisation is carried out under stringent hybridisation and washing conditions (e.g. hybridisation at 65° C. in a buffer containing 5 ⁇ SSC; washing at 42° C. with 0.2 ⁇ SSC/0.1% SDS).
  • stringent hybridisation and washing conditions e.g. hybridisation at 65° C. in a buffer containing 5 ⁇ SSC; washing at 42° C. with 0.2 ⁇ SSC/0.1% SDS.
  • stringent hybridisation and washing conditions e.g. hybridisation at 65° C. in a buffer containing 5 ⁇ SSC; washing at 42° C. with 0.2 ⁇ SSC/0.1% SDS.
  • sequence region 1 includes all or at least parts of the sequence region between nucleotide position 1 bp and 1578 bp. This corresponds to sequence region 5′ of the TE-00 sequence (SEQ ID No. 2).
  • the fragment TE-08 (SEQ ID No. 10) is also preferrred.
  • variant refers to the expression vectors used in the particular transfection mixture. These include both the base vectors (pTE4/MCP-1 or pTE5/MCP-1) or the base vector combination (pBING-LC+pBID-HC) and also the base vectors which contain one or more TE elements in different positions, combinations and orientations.
  • the term “orientation” refers to the arrangement in relation to the gene of interest.
  • the sequence given under SEQ ID No. 1 represents a genome sequence which is positioned 5′ from the region coding for the ubiquitin/S27a gene, which is also referred to as “upstream”.
  • the TE element is in the direct orientation when the sequence shown in SEQ ID No. 1, or any desired part, fragment, region or derivative thereof, is present in the expression vector on the same DNA strand as the start codon of the gene of interest. If, by contrast, the sequence complementary to SEQ ID No. 1, or any desired part, fragment, region or derivative thereof, is present in the expression vector on the same DNA strand as the start codon of the gene of interest, then the TE element is in a “reverse orientation”. Unless stated otherwise, when a TE element is mentioned, both orientations are always included/meant, i.e. both direct and reverse.
  • chromosomal integration is meant the integration of any desired nucleic acid sequence into the genome, i.e. into the chromosomes, of a cell, this integration optionally being into one or more chromosomes in any desired number, position and orientation.
  • chromosomal integration also includes the integration of any desired nucleic acid sequence into synthetic, artificial or mini-chromosomes.
  • the gene of interest contained in the expression vector according to the invention comprises a nucleotide sequence of any length which codes for a product of interest.
  • the gene product or “product of interest” is generally a protein, polypeptide, peptide or fragment or derivative thereof. However, it may also be RNA or antisense RNA.
  • the gene of interest may be present in its full length, in shortened form, as a fusion gene or as a labelled gene. It may be genomic DNA or preferably cDNA or corresponding fragments or fusions.
  • the gene of interest may be the native gene sequence, or it may be mutated or otherwise modified. Such modifications include codon optimisations for adapting to a particular host cell and humanisation.
  • the gene of interest may, for example, code for a secreted, cytoplasmic, nuclear-located, membrane-bound or cell surface-bound polypeptide.
  • nucleotide sequence indicates an oligonucleotide, nucleotides, polynucleotides and fragments thereof as well as DNA or RNA of genomic or synthetic origin which occur as single or double strands and can represent the coding or non-coding strand of a gene.
  • Nucleic acid sequences may be modified using standard techniques such as site-specific mutagenesis or PCR-mediated mutagenesis (e.g. described in Sambrook et al., 1989 or Ausubel et al., 1994).
  • coding is meant the property or capacity of a specific sequence of nucleotides in a nucleic acid, for example a gene in a chromosome or an mRNA, to act as a matrix for the synthesis of other polymers and macromolecules such as for example rRNA, tRNA, mRNA, other RNA molecules, cDNA or polypeptides in a biological process. Accordingly, a gene codes for a protein if the desired protein is produced in a cell or another biological system by transcription and subsequent translation of the mRNA. Both the coding strand whose nucleotide sequence is identical to the mRNA sequence and is normally also given in sequence databanks, e.g.
  • nucleic acid which codes for a protein also includes nucleic acids which have a different order of nucleotide sequence on the basis of the degenerate genetic code but result in the same amino acid sequence of the protein. Nucleic acid sequences which code for proteins may also contain introns.
  • cDNA denotes deoxyribonucleic acids which are prepared by reverse transcription and synthesis of the second DNA strand from a mRNA or other RNA produced from a gene. If the cDNA is present as a double stranded DNA molecule it contains both a coding and a non-coding strand.
  • intron denotes non-coding nucleotide sequences of any length. They occur naturally in numerous eukaryotic genes and are eliminated from a previously transcribed mRNA precursor by a process known as splicing. This requires precise excision of the intron at the 5′ and 3′ ends and correct joining of the resulting mRNA ends so as to produce a mature processed mRNA with the correct reading frame for successful protein synthesis. Many of the splice donor and splice acceptor sites involved in this splicing process, i.e. the sequences located directly at the exon-intron or intron-exon interfaces, have been characterised by now. For an overview see Ohshima et al., 1987.
  • Proteins/polypeptides with a biopharmaceutical significance include for example antibodies, enzymes, cytokines, lymphokines, adhesion molecules, receptors and the derivatives or fragments thereof, but are not restricted thereto. Generally, all polypeptides which act as agonists or antagonists and/or have therapeutic or diagnostic applications may be used. Other proteins of interest are, for example, proteins/polypeptides, which are used to change the properties of host cells within the scope of so-called “Cell Engineering”, such as e.g. anti-apoptotic proteins, chaperones, metabolic enzymes, glycosylation enzymes and the derivatives or fragments thereof, but are not restricted thereto.
  • Cell Engineering such as e.g. anti-apoptotic proteins, chaperones, metabolic enzymes, glycosylation enzymes and the derivatives or fragments thereof, but are not restricted thereto.
  • polypeptides is used for amino acid sequences or proteins and refers to polymers of amino acids of any length. This term also includes proteins which have been modified post-translationally by reactions such as glycosylation, phosphorylation, acetylation or protein processing.
  • the structure of the polypeptide may be modified, for example, by substitutions, deletions or insertions of amino acids and fusion with other proteins while retaining its biological activity.
  • the polypeptides may multimerise and form homo- and heteromers.
  • therapeutic proteins are insulin, insulin-like growth factor, human growth hormone (hGH) and other growth factors, receptors, tissue plasminogen activator (tPA), erythropoietin (EPO), cytokines, e.g. interleukines (IL) such as IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, interferon (IFN)-alpha, -beta, -gamma, -omega or -tau, tumour necrosis factor (TNF) such as TNF-alpha, beta or gamma, TRAIL, G-CSF, GM-CSF, M-CSF, MCP-1 and VEGF.
  • IL interleukines
  • IFN interferon
  • TNF tumour necrosis factor
  • antibodies are monoclonal, polyclonal, multispecific and single chain antibodies and fragments thereof such as for example Fab, Fab′, F(ab′) 2 , Fc and Fc′ fragments, light (L) and heavy (H) immunoglobulin chains and the constant, variable or hypervariable regions thereof as well as Fv and Fd fragments (Chamov et al., 1999).
  • the antibodies may be of human or non-human origin. Humanised and chimeric antibodies are also possible.
  • Fab fragments consist of the variable regions of both chains which are held together by the adjacent constant regions. They may be produced for example from conventional antibodies by treating with a protease such as papain or by DNA cloning. Other antibody fragments are F(ab′) 2 fragments which can be produced by proteolytic digestion with pepsin.
  • the variable regions of the heavy and light chains are often joined together by means of a short peptide fragment of about 10 to 30 amino acids, preferably 15 amino acids. This produces a single polypeptide chain in which VH and VL are joined together by a peptide linker.
  • Such antibody fragments are also referred to as single chain Fv fragments (scFv). Examples of scFv antibodies are known and described, cf. for example Huston et al., 1988.
  • multimeric scFv derivatives In past years various strategies have been developed for producing multimeric scFv derivatives. The intention is to produce recombinant antibodies with improved pharmacokinetic properties and increased binding avidity. In order to achieve the multimerisation of the scFv fragments they are produced as fusion proteins with multimerisation domains.
  • the multimerisation domains may be, for example, the CH3 region of an IgG or helix structures (“coiled coil structures”) such as the Leucine Zipper domains.
  • the interactions between the VH and VL regions of the scFv fragment are used for multimerisation (e.g. dia, tri- and pentabodies).
  • diabody is used in the art to denote a bivalent homodimeric scFv derivative. Shortening the peptide linker in the scFv molecule to 5 to 10 amino acids results in the formation of homodimers by superimposing VHNVL chains.
  • the diabodies may additionally be stabilised by inserted disulphite bridges. Examples of diabodies can be found in the literature, e.g. in Perisic et al., 1994.
  • minibody is used in the art to denote a bivalent homodimeric scFv derivative. It consists of a fusion protein which contains the CH3 region of an immunoglobulin, preferably IgG, most preferably IgG1, as dimerisation region. This connects the scFv fragments by means of a hinge region, also of IgG, and a linker region. Examples of such minibodies are described by Hu et al., 1996.
  • trimers are used in the art to denote a trivalent homotrimeric scFv derivative (Kortt et al., 1997). The direct fusion of VH-VL without the use of a linker sequence leads to the formation of trimers.
  • fragments known in the art as mini antibodies which have a bi, tri- or tetravalent structure are also derivatives of scFv fragments.
  • the multimerisation is achieved by means of di-, tri- or tetrameric coiled coil structures (Pack et al., 1993 and 1995; Lovejoy et al., 1993).
  • the expression vector according to the invention contains a gene coding for a fluorescent protein, functionally linked to the gene of interest.
  • both genes are transcribed under the control of a single heterologous promoter so that the protein/ product of interest and the fluorescent protein are coded by a bicistronic mRNA. This makes it possible to identify cells which produce the protein/product of interest in large amounts, by means of the expression rate of the fluorescent protein.
  • the transcription of the gene coding for the fluorescent protein may take place under the control of its own promoter.
  • the fluorescent protein may be, for example, a green, bluish-green, blue, yellow or other coloured fluorescent protein.
  • GFP green fluorescent protein
  • fluorescent proteins and genes coding for them are described in WO 00/34318, WO 00/34326, WO 00/34526 and WO 01/27150 which are incorporated herein by reference.
  • These fluorescent proteins are fluorophores of non-bioluminescent organisms of the species Anthozoa, for example Anemonia majano, Clavularia sp., Zoanthus sp. I, Zoanthus sp. II, Discosoma striata, Discosoma sp. “red”, Discosoma sp. “green”, Discosoma sp. “Magenta”, Anemonia sulcata, Aequorea coerulescens.
  • the fluorescent proteins used according to the invention contain in addition to the wild-type proteins natural or genetically engineered mutants and variants, fragments, derivatives or variants thereof which have for example been fused with other proteins or peptides.
  • the mutations used may for example alter the excitation or emission spectrum, the formation of chromophores, the extinction coefficient or the stability of the protein.
  • the expression in mammalian cells or other species can be improved by codon optimisation.
  • the fluorescent protein may also be used in fusion with a selectable marker, preferably an amplifiable selectable marker such as dihydrofolate reductase (DHFR).
  • DHFR dihydrofolate reductase
  • the fluorescence emitted by the fluorescent proteins makes it possible to detect the proteins, e.g. by throughflow cytometry with a fluorescence-activated cell sorter (FACS) or by fluorescence microscopy.
  • FACS fluorescence-activated cell sorter
  • the expression vector contains at least one heterologous promoter which allows expression of the gene of interest and preferably also of the fluorescent protein.
  • promoter denotes a polynucleotide sequence which allows and controls the transcription of the genes or sequences functionally connected therewith.
  • a promoter contains recognition sequences for binding RNA polymerase and the initiation site for transcription (transcription initiation site).
  • a suitable functional promoter In order to express a desired sequence in a certain cell type or a host cell a suitable functional promoter must be chosen.
  • the skilled man will be familiar with a variety of promoters from various sources, including constitutive, inducible and repressible promoters. They are deposited in databanks such as GenBank, for example, and may be obtained as separate elements or elements cloned within polynucleotide sequences from commercial or individual sources.
  • inducible promoters the activity of the promoter may be reduced or increased in response to a signal.
  • an inducible promoter is the tetracycline (tet) promoter. This contains tetracycline operator sequences (tetO) which can be induced by a tetracycline-regulated transactivator protein (tTA). In the presence of tetracycline the binding of tTA to tetO is inhibited.
  • tetO tetracycline operator sequences
  • tTA tetracycline-regulated transactivator protein
  • tTA tetracycline-regulated transactivator protein
  • jun, fos, metallothionein and heat shock promoter see also Sambrook et al., 1989; Gossen et al., 1994).
  • promoters which are particularly suitable for high expression in eukaryotes, there are for example the ubiquitin/S27a promoter of the hamster (WO 97/15664), SV 40 early promoter, adenovirus major late promoter, mouse metallothionein-I promoter, the long terminal repeat region of Rous Sarcoma Virus, the early promoter of human Cytomegalovirus.
  • promoters which are particularly suitable for high expression in eukaryotes, there are for example the ubiquitin/S27a promoter of the hamster (WO 97/15664), SV 40 early promoter, adenovirus major late promoter, mouse metallothionein-I promoter, the long terminal repeat region of Rous Sarcoma Virus, the early promoter of human Cytomegalovirus.
  • heterologous mammalian promoters are the actin, immunoglobulin or heat shock promoter(s).
  • a corresponding heterologous promoter can be functionally connected to other regulatory sequences in order to increase/regulate the transcription activity in an expression cassette.
  • the promoter may be functionally linked to enhancer sequences in order to increase the transcriptional activity.
  • one or more enhancers and/or several copies of an enhancer sequence may be used, e.g. a CMV or SV40 enhancer.
  • an expression vector according to the invention in another embodiment, contains one or more enhancers/ enhancer sequences, preferably a CMV or SV40 enhancer.
  • enhancer denotes a polynucleotide sequence which in the cis location acts on the activity of a promoter and thus stimulates the transcription of a gene functionally connected to this promoter. Unlike promoters the effect of enhancers is independent of position and orientation and they can therefore be positioned in front of or behind a transcription unit, within an intron or even within the coding region. The enhancer may be located both in the immediate vicinity of the transcription unit and at a considerable distance from the promoter. It is also possible to have a physical and functional overlap with the promoter. The skilled man will be aware of a number of enhancers from various sources (and deposited in databanks such as GenBank, e.g.
  • SV40 enhancers CMV enhancers, polyoma enhancers, adenovirus enhancers
  • CMV enhancers CMV enhancers
  • polyoma enhancers SV40 enhancers
  • adenovirus enhancers which are available as independent elements or elements cloned within polynucleotide sequences (e.g. deposited at the ATCC or from commercial and individual sources).
  • a number of promoter sequences also contain enhancer sequences such as the frequently used CMV promoter.
  • the human CMV enhancer is one of the strongest enhancers identified hitherto.
  • an inducible enhancer is the metallothionein enhancer, which can be stimulated by glucocorticoids or heavy metals.
  • the promoter sequences may also be combined with regulatory sequences which allow control/regulation of the transcription activity.
  • the promoter can be made repressible/ inducible. This can be done for example by linking to sequences which are binding sites for up- or down-regulating transcription factors.
  • the above mentioned transcription factor Sp1 for example, has a positive effect on the transcription activity.
  • Another example is the binding site for the activator protein AP1, which may act both positively and negatively on transcription.
  • the activity of AP1 can be controlled by all kinds of factors such as, for example, growth factors, cytokines and serum (Faisst et al., 1992 and references therein).
  • the transcription efficiency can also be increased by changing the promoter sequence by the mutation (substitution, insertion or deletion) of one, two, three or more bases and then determining, in a reporter gene assay, whether this has increased the promoter activity.
  • the additional regulatory elements include heterologous promoters, enhancers, termination and polyadenylation signals and other expression control elements. Both inducible and constitutively regulatory sequences are known for the various cell types.
  • Transcription-regulatory elements generally comprise a promoter upstream of the gene sequence to be expressed, transcription initiation and termination sites and a polyadenylation signal.
  • transcription initiation site refers to a nucleic acid in the construct which corresponds to the first nucleic acid which is incorporated in the primary transcript, i.e. the mRNA precursor.
  • the transcription initiation site may overlap with the promoter sequences.
  • transcription termination site refers to a nucleotide sequence which is normally at the 3′ end of the gene of interest or of the gene section which is to be transcribed, and which brings about the termination of transcription by RNA polymerase.
  • the “polyadenylation signal” is a signal sequence which causes cleavage at a specific site at the 3′ end of the eukaryotic mRNA and post-transcriptional incorporation of a sequence of about 100-200 adenine nucleotides (polyA tail) at the cleaved 3′ end.
  • the polyadenylation signal comprises the sequence AATAAA about 10-30 nucleotides upstream of the cleavage site and a sequence located downstream.
  • Various polyadenylation elements are known such as tk polyA, SV40 late and early polyA or BGH polyA (described for example in U.S. Pat. No. 5,122,458).
  • Translation regulatory elements comprise a translation initiation site (AUG), a stop codon and a polyA signal for each polypeptide to be expressed.
  • AUG translation initiation site
  • stop codon a polyA signal for each polypeptide to be expressed.
  • ribosomal consensus binding sites may alternatively be inserted immediately upstream of the start codon.
  • the gene of interest usually contains a signal sequence which codes for a signal precursor peptide which transports the synthesised polypeptide to and through the ER membrane.
  • the signal sequence is often but not always located at the amino terminus of the secreted protein and is cleaved by signal peptidases after the protein has been filtered through the ER membrane.
  • the gene sequence will usually but not necessarily contain its own signal sequence. If the native signal sequence is not present a heterologous signal sequence may be introduced in known manner. Numerous signal sequences of this kind are known to the skilled man and deposited in sequence databanks such as GenBank and EMBL.
  • the IRES element comprises a sequence which functionally activates the translation initiation independently of a 5′-terminal methylguanosinium cap (CAP structure) and the upstream gene and in an animal cell allows the translation of two cistrons (open reading frames) from a single transcript.
  • the IRES element provides an independent ribosomal entry site for the translation of the open reading frame located immediately downstream.
  • bacterial mRNA which may be multicistronic, i.e. it may code for numerous different polypeptides or products which are translated one after the other by the mRNA, the majority of mRNAs from animal cells are monocistronic and code for only one protein or product.
  • a multicistronic transcript in a eukaryotic cell the translation would be initiated from the translation initiation site which was closest upstream and would be stopped by the first stop codon, after which the transcript would be released from the ribosome. Thus, only the first polypeptide or product coded by the mRNA would be produced during translation.
  • a multicistronic transcript with an IRES element which is functionally linked to the second or subsequent open reading frame in the transcript allows subsequent translation of the open reading frame located downstream thereof, so that two or more polypeptides or products coded by the same transcript are produced in the eukaryotic cell.
  • the IRES element may be of various lengths and various origins and may originate, for example, from the encephalomyocarditis virus (EMCV) or other Picorna viruses.
  • EMCV encephalomyocarditis virus
  • IRES sequences and their use in the construction of vectors are described in the literature, cf. for example Pelletier et al., 1988; Jang et al., 1989; Davies et al., 1992; Adam et al., 1991; Morgan et al., 1992; Sugimoto et al., 1994; Ramesh et al., 1996; Mosser et al., 1997.
  • the gene sequence located downstream is functionally linked to the 3′ end of the IRES element, i.e. the spacing is selected so that the expression of the gene is unaffected or only marginally affected or has sufficient expression for the intended purpose.
  • the optimum permissible distance between the IRES element and the start codon of the gene located downstream thereof for sufficient expression can be determined by simple experiments by varying the spacing and determining the expression rate as a function of the spacing using reporter gene assays.
  • the expression vector according to the invention contains the ubiquitin/S27a promoter of the hamster, preferably functionally linked to the gene of interest and even more preferably functionally linked to the gene of interest and the gene which codes for a fluorescent protein or a selectable marker.
  • the ubiquitin/S27a promoter of the hamster is a powerful homologous promoter which is described in WO 97/15664.
  • a promoter preferably has at least one of the following features: GC-rich sequence area, Sp1 binding site, polypyrimidine element, absence of a TATA box.
  • a promoter which has an Sp1 binding site but no TATA box is particularly preferred.
  • it is an inducible promoter, particularly a promoter which is activated by the removal of serum.
  • a particularly advantageous embodiment is a promoter with a nucleotide sequence as contained in FIG. 5 of WO 97/15664. Particularly preferred are promoter sequences which contain the sequence from position ⁇ 161 to ⁇ 45 of FIG. 5 .
  • the promoters used in the examples of the present patent specification each contain a DNA molecule with a sequence which corresponds to the fragment ⁇ 372 to +111 from FIG. 5 of WO 97/15664 and represents the preferred promoter, i.e a preferred promoter should incorporate this sequence region.
  • the expression vector according to the invention may theoretically be prepared by conventional methods known in the art, as described by Sambrook et al. (1989), for example.
  • Sambrook also describes the functional components of a vector, e.g. suitable promoters (in addition to the hamster ubiquitin/S27a promoter), enhancers, termination and polyadenylation signals, antibiotic resistance genes, selectable markers, replication starting points and splicing signals.
  • suitable promoters in addition to the hamster ubiquitin/S27a promoter
  • enhancers in addition to the hamster ubiquitin/S27a promoter
  • termination and polyadenylation signals e.g. suitable antibiotic resistance genes, selectable markers, replication starting points and splicing signals.
  • Conventional cloning vectors may be used to produce them, e.g.
  • plasmids bacteriophages, phagemids, cosmids or viral vectors such as baculovirus, retroviruses, adenoviruses, adeno-associated viruses and herpes simplex virus, as well as synthetic or artificial chromosomes/mini chromosomes.
  • the eukaryotic expression vectors typically also contain prokaryotic sequences such as, for example, replication origin and antibiotic resistance genes which allow replication and selection of the vector in bacteria.
  • a number of eukaryotic expression vectors which contain multiple cloning sites for the introduction of a polynucleotide sequence are known and some may be obtained commercially from various companies such as Stratagene, La Jolla, Calif., USA; Invitrogen, Carlsbad, Calif., USA; Promega, Madison, Wis., USA or BD Biosciences Clontech, Palo Alto, Calif., USA.
  • the heterologous promoter, the gene (or genes) of interest, selectable markers and optionally the gene coding for a fluorescent protein, additional regulatory elements such as the internal ribosomal entry site (IRES), enhancers, a polyadenylation signal and other cis-active elements such as TE elements, for example, are introduced into the expression vector in a manner familiar to those skilled in the art.
  • An expression vector according to the invention contains, at the minimum, a heterologous promoter, the gene of interest and a TE element.
  • the expression vector also contains a gene coding for a fluorescent protein. It is particularly preferred according to the invention to use a ubiquitin/S27a promoter as heterologous promoter.
  • Particularly preferred is an expression vector in which the heterologous promoter, preferably a ubiquitin/S27a promoter, the gene of interest and a TE element are functionally linked together or are functionally linked.
  • a promoter/enhancer, a promoter/TE element or a promoter/enhancer/TE element is functionally linked to a coding gene sequence if it is able to control or modulate the transcription of the linked gene sequence in the cis position.
  • functionally linked DNA sequences are close together and, if two coding gene sequences are linked or in the case of a secretion signal sequence, in the same reading frame.
  • a functionally linked promoter is generally located upstream of the coding gene sequence it does not necessarily have to be close to it.
  • Enhancers need not be close by either, provided that they assist the transcription or expression of the gene sequence. For this purpose they may be both upstream and downstream of the gene sequence, possibly at some distance from it.
  • a polyadenylation site is functionally linked to a gene sequence if it is positioned at the 3′ end of the gene sequence in such a way that the transcription progresses via the coding sequence to the polyadenylation signal. Linking may take place according to conventional recombinant methods, e.g. by the PCR technique, by ligation at suitable restriction cutting sites or by splicing. If no suitable restriction cutting sites are available synthetic oligonucleotide linkers or adaptors may be used in a manner known per se.
  • the heterologous promoter preferably a ubiquitin/S27a promoter or CMV promoter
  • the gene of interest and the gene coding for a fluorescent protein are functionally linked together.
  • the functional linking takes place via an IRES element, so that a bicistronic mRNA is synthesised from both genes.
  • the expression vector according to the invention may additionally contain enhancer elements and/or TE elements which act functionally on one or more promoters.
  • an enhancer element e.g. an SV40 enhancer or a CMV enhancer element
  • a TE element e.g. an SV40 enhancer or a CMV enhancer element
  • the expression of the genes within an expression vector may take place starting from one or more transcription units.
  • transcription unit is defined as a region which contains one or more genes to be transcribed.
  • the genes within a transcription unit are functionally linked to one another in such a way that all the genes within such a unit are under the transcriptional control of the same promoter, promoter/enhancer or promoter/enhancer/TE element.
  • each transcription unit contains the regulatory elements which are necessary for the transcription and translation of the gene sequences contained therein.
  • Each transcription unit may contain the same or different regulatory elements. IRES elements or introns may be used for the functional linking of the genes within a transcription unit.
  • the expression vector may contain a single transcription unit for expressing the gene (or genes) of interest, the selectable marker and optionally the gene which codes for the fluorescent protein. Alternatively, these genes may also be arranged in two or more transcription units. Various combinations of the genes within a transcription unit are possible. In another embodiment of the present invention more than one expression vector consisting of one, two or more transcription units may be inserted in a host cell by cotransfection or in successive transfections in any desired order. Any combination of regulatory elements and genes on each vector can be selected provided that adequate expression of the transcription units is ensured. If necessary, other regulatory elements, such as TE elements, and genes, e.g. additional genes of interest or selectable markers, may be positioned on the expression vectors.
  • Also preferred according to the invention are those expression vectors which contain one or more TE elements and instead of the gene of interest have only a multiple cloning site which allows the cloning of the gene of interest via recognition sequences for restriction endonucleases.
  • recognition sequences for all kinds of restriction endonucleases as well as the associated restriction endonucleases are known from the prior art.
  • sequences are used which consist of at least six nucleotides as recognition sequence. A list of suitable recognition sequences can be found for example in Sambrook et al., 1989.
  • Also preferred according to the invention are those expression vectors which instead of the gene of interest have only a multiple cloning site which allows the cloning of the gene of interest via recognition sequences for restriction endonucleases and which moreover have one or more, preferably multiple cloning sites at different positions of the expression vector, which additionally makes it possible to clone TE elements via recognition sequences for restriction endonucleases.
  • Numerous recognition sequences for all kinds of restriction endonucleases as well as the associated restriction endonucleases are known from the prior art.
  • sequences are used which consist of at least six nucleotides as recognition sequence. A list of suitable recognition sequences can be found for example in Sambrook et al., 1989.
  • eukaryotic host cells preferably mammalian cells and more particularly rodent cells such as mouse, rat and hamster cell lines.
  • rodent cells such as mouse, rat and hamster cell lines.
  • Preferred host cells for the purposes of the invention are hamster cells such as BHK21, BHK TK ⁇ , CHO, CHO-K1, CHO-DUKX, CHO-DUKX B1 and CHO-DG44 cells or derivatives/descendants of these cell lines.
  • Particularly preferred are CHO-DG44, CHO-DUKX, CHO-K1 and BHK21 cells, particularly CHO-DG44 and CHO-DUKX cells.
  • myeloma cells from the mouse preferably NSO and Sp2/0 cells and derivatives/descendants of these cell lines.
  • hamster and mouse cells which can be used according to the invention are given in Table 1 that follows.
  • other mammalian cells including but not restricted to cell lines of humans, mice, rats, monkeys, rodents, or eukaryotic cells, including but not restricted to yeast, insect, bird and plant cells, may also be used as host cells for the production of biopharmaceutical proteins.
  • transfection of the eukaryotic host cells with a polynucleotide or one of the expression vectors according to the invention is carried out by conventional methods (Sambrook et al. 1989; Ausubel et al., 1994). Suitable methods of transfection include for example liposome-mediated transfection, calcium phosphate coprecipitation, electroporation, polycation- (e.g. DEAE dextran)-mediated transfection, protoplast fusion, microinjection and viral infections.
  • stable transfection is preferably carried out in which the constructs are either integrated into the genome of the host cell or an artificial chromosome/minichromosome, or are episomally contained in stable manner in the host cell.
  • heterologous sequence or “heterologous gene” in relation to the host cell. This applies even if the sequence to be introduced or the gene to be introduced is identical to an endogenous sequence or an endogenous gene of the host cell.
  • a hamster actin gene introduced into a hamster host cell is by definition a heterologous gene.
  • recombinant mammalian cells preferably rodent cells, most preferably hamster cells such as CHO or BHK cells which have been transfected with one of the expression vectors according to the invention described herein are preferred.
  • mAb monoclonal antibodies
  • the transfection of suitable host cells can theoretically be carried out by two different methods.
  • mAb's of this kind are composed of a number of subunits, the heavy and light chains. Genes coding for these subunits may be accommodated in independent or multicistronic transcription units on a single plasmid with which the host cell is then transfected. This is intended to secure the stoichiometric representation of the genes after integration into the genome of the host cell.
  • independent transcription units it must hereby be ensured that the mRNAs which encode the different proteins display the same stability and transcriptional and translational efficiency.
  • the expression of the genes take place within a multicistronic transcription unit by means of a single promoter and only one transcript is formed.
  • IRES elements By using IRES elements, a highly efficient internal translation initiation of the genes is obtained in the second and subsequent cistrons.
  • the expression rates for these cistrons are lower than that of the first cistron, the translation initiation of which, by means of a so-called “cap”-dependent pre-initiation complex, is substantially more efficient than IRES-dependent translation initiation.
  • additional inter-cistronic elements may be introduced, for example, which ensure uniform expression rates in conjunction with the IRES elements (WO 94/05785).
  • Another possible way of simultaneously producing a number of heterologous proteins is cotransfection, in which the genes are separately integrated in different expression vectors.
  • This has the advantage that certain proportions of genes and gene products with one another can be adjusted, thereby balancing out any differences in the mRNA stability and in the efficiency of transcription and translation.
  • the expression vectors are more stable because of their small size and are easier to handle both during cloning and during transfection.
  • the host cells are additionally transfected, preferably cotransfected, with one or more vectors having genes which code for one or more other proteins of interest.
  • the other vector or vectors used for the cotransfection code for example, for the other protein or proteins of interest under the control of the same promoter, preferably under the control of the same promoter/enhancer combination or, particularly preferably, under the control of the same promoter/enhancer/TE element combination or under the control of the same promoter/enhancer combination with different TE elements and for at least one selectable marker, e.g. dihydrofolate reductase.
  • the vectors used for the transfection may contain one or more TE-elements in any combination, position and orientation.
  • the host cells are co-transfected with at least two eukaryotic expression vectors, at least one of the two vectors containing at least one gene which codes for at least the protein of interest, while the other vector contains one or more nucleic acids according to the invention in any combination, position and orientation, and optionally also codes for at least one gene of interest, and these nucleic acids according to the invention impart their transcription- or expression-enhancing activity to the genes of interest which are located on the other co-transfected vector, by co-integration with the other vector.
  • the host cells are preferably established, adapted and cultivated under serum-free conditions, optionally in media which are free from animal proteins/peptides.
  • media include Ham's F12 (Sigma, Deisenhofen, Del.), RPMI-1640 (Sigma), Dulbecco's Modified Eagle's Medium (DMEM; Sigma), Minimal Essential Medium (MEM; Sigma), Iscove's Modified Dulbecco's Medium (IMDM; Sigma), CD-CHO (Invitrogen, Carlsbad, Calif., USA), CHO-S-SFMII (Invitrogen), serum-free CHO-Medium (Sigma) and protein-free CHO-Medium (Sigma).
  • Each of these media may optionally be supplemented with various compounds, e.g. hormones and/or other growth factors (e.g. insulin, transferrin, epidermal growth factor, insulin-like growth factor), salts (e.g. sodium chloride, calcium, magnesium, phosphate), buffers (e.g. HEPES), nucleosides (e.g. adenosine, thymidine), glutamine, glucose or other equivalent nutrients, antibiotics and/or trace elements.
  • growth factors e.g. insulin, transferrin, epidermal growth factor, insulin-like growth factor
  • salts e.g. sodium chloride, calcium, magnesium, phosphate
  • buffers e.g. HEPES
  • nucleosides e.g. adenosine, thymidine
  • glutamine glucose or other equivalent nutrients
  • antibiotics and/or trace elements e.g., antibiotics and/or trace elements.
  • serum-free media are preferred according to the invention, the host cells may also be
  • selecting agent refers to a substance which affects the growth or survival of host cells with a deficiency for the selectable marker gene in question.
  • geneticin G4108 is preferably used as the medium additive for the selection of heterologous host cells which carry a wild-type or preferably a modified neomycin phosphotransferase gene.
  • G418 concentrations of between 100 and 800 ⁇ g/ml of medium are used, most preferably 200 to 400 ⁇ g G418/ml of medium. If the host cells are to be transfected with a number of expression vectors, e.g. if several genes of interest are to be separately introduced into the host cell, they generally have different selectable marker genes.
  • a selectable marker gene is a gene which allows the specific selection of cells which contain this gene by the addition of a corresponding selecting agent to the cultivation medium.
  • an antibiotic resistance gene may be used as a positive selectable marker. Only cells which have been transformed with this gene are able to grow in the presence of the corresponding antibiotic and are thus selected. Untransformed cells, on the other hand, are unable to grow or survive under these selection conditions.
  • the selectable markers used in this invention include genetically modified mutants and variants, fragments, functional equivalents, derivatives, homologues and fusions with other proteins or peptides, provided that the selectable marker retains its selective qualities.
  • Such derivatives display considerable homology in the amino acid sequence in the regions or domains which are deemed to be selective.
  • the literature describes a large number of selectable marker genes including bifunctional (positive/negative) markers (see for example WO 92/08796 and WO 94/28143).
  • selectable markers which are usually used in eukaryotic cells include the genes for aminoglycoside phosphotransferase (APH), hygromycine phosphostransferase (HYG), dihydrofolate reductase (DHFR), thymidine kinase (TK), glutamine synthetase, asparagin synthetase and genes which confer resistance to neomycin (G418), puromycin, histidinol D, bleomycin, phleomycin and zeocin.
  • APH aminoglycoside phosphotransferase
  • HAG hygromycine phosphostransferase
  • DHFR dihydrofolate reductase
  • TK thymidine kinase
  • glutamine synthetase glutamine synthetase
  • asparagin synthetase genes which confer resistance to neomycin (G418), puro
  • modified neomycin-phosphotransferase covers all the mutants described in WO2004/050884, particularly the mutant D227G (Asp227Gly), which is characterised by the substitution of aspartic acid (Asp, D) for glycine (Gly, G) at amino acid position 227 and particularly preferably the mutant F2401 (Phe240Ile), which is characterised by the substitution of phenylalanine (Phe, F) for isoleucine (Ile, J) at amino acid position 240.
  • the present invention therefore includes a method of preparing and selecting recombinant mammalian cells which comprises the following steps: (i) transfecting the host cells with genes which code for at least one protein/product of interest and a neomycin-phosphotransferase, preferably modified, wherein in order to enhance the transcription or expression at least the gene (or genes) of interest is functionally linked to at least one TE element; (ii) cultivating the cells under conditions that enable expression of the different genes; and (iii) selecting these co-integrated genes by cultivating the cells in the presence of a selecting agent such as e.g. G418.
  • the transfected cells are cultivated in medium in the absence of serum.
  • the concentration of G418 is at least 200 ⁇ g/mL. However, the concentration may also be at least 400 ⁇ g/mL.
  • the cells according to the invention may optionally also be subjected to one or more gene amplification steps in which they are cultivated in the presence of a selecting agent which leads to amplification of an amplifiable selectable marker gene.
  • the host cells are additionally transfected with a gene which codes for an amplifiable selectable marker. It is conceivable for the gene which codes for an amplifiable selectable marker to be present on one of the expression vectors according to the invention or to be introduced into the host cell by means of another vector.
  • the amplifiable selectable marker gene usually codes for an enzyme which is needed for the growth of eukaryotic cells under certain cultivation conditions.
  • the amplifiable selectable marker gene may code for dihydrofolate reductase (DHFR).
  • DHFR dihydrofolate reductase
  • the gene is amplified if a host cell transfected therewith is cultivated in the presence of the selecting agent methotrexate (MTX).
  • MTX methotrexate
  • Table 2 gives examples of other amplifiable selectable marker genes and the associated selecting agents which may be used according to the invention, which are described in an overview by Kaufman, Methods in Enzymology, 185:537-566 (1990).
  • J03398 (mouse) adriamycin, vincristin, colchicine ribonucleotide reductase M124223, K02927 aphidicoline (mouse) glutamine-synthetase AF150961 (hamster) methionine sulphoximine U09114, M60803 (mouse) (MSX) M29579 (rat) asparagine-synthetase M27838 (hamster) ⁇ -aspartylhydroxamate, M27396 (human) albizziin, 5′azacytidine U38940 (mouse) U07202 (rat) argininosuccinate-synthetase X01630 (human) canavanin M31690 (mouse) M26198 (bovine) ornithine-decarboxylase M34158 (human) ⁇ -difluoromethylornithine J0
  • the amplifiable selectable marker gene used is preferably a gene which codes for a polypeptide with the function of DHFR, e.g. for DHFR or a fusion protein from the fluorescent protein and DHFR.
  • DHFR is necessary for the biosynthesis of purines. Cells which lack the DHFR genes cannot grow in purine-deficient medium.
  • the DHFR gene is therefore a useful selectable marker for selecting and amplifying genes in cells cultivated in purine-free medium.
  • the selecting medium used in conjunction with the DHFR gene is methotrexate (MTX).
  • Mammalian cells preferably mouse myeloma and hamster cells, are preferred host cells for the use of DHFR as an amplifiable selectable marker.
  • the cell lines CHO-DUKX (ATCC CRL-9096) and CHO-GD44 (Urlaub et al., 1983) are particularly preferred as they have no DHFR activity of their own, as a result of mutation.
  • DHFR-induced amplification in other cell types as well which have their own endogenous DHFR activity, it is possible to use in the transfection process a mutated DHFR gene which codes for a protein with reduced sensitivity to methotrexate (Simonson et al., 1983; Wigler et al., 1980; Haber et al., 1982).
  • the DHFR marker is particularly suitable for the selection and subsequent amplification when using DHFR-negative basic cells such as CHO-DG44 or CHO-DUKX, as these cells do not express endogenous DHFR and therefore do not grow in purine-free medium. Consequently, the DHFR gene may be used here as a dominant selectable marker and the transformed cells are selected in hypoxanthine/thymidine-free medium.
  • the present invention therefore includes a method of preparing and selecting recombinant mammalian cells which comprises the following steps: (i) transfecting the host cells with genes which code for at least one protein/product of interest and the amplifiable selectable marker DHFR, wherein in order to enhance the transcription or expression at least the gene (or genes) of interest is functionally linked to at least one TE element; (ii) cultivating the cells under conditions that enable expression of the different genes; and (iii) amplifying these co-integrated genes by cultivating the cells in the presence of a selecting agent which allows the amplification of at least the amplifiable selectable marker gene, such as methotrexate.
  • a selecting agent which allows the amplification of at least the amplifiable selectable marker gene, such as methotrexate.
  • the transfected cells are cultivated in hypoxanthine/thymidine-free medium in the absence of serum and with the addition of increasing concentrations of MTX.
  • concentration of MTX in the first amplification step is at least 100 nM.
  • concentration of MTX may, however, also be at least 250 nM and may be increased step by step to 1 ⁇ M. In individual cases concentrations of over 1 ⁇ M may also be used, e.g. 2 ⁇ M.
  • the present invention also includes a method of preparing and selecting recombinant mammalian cells which comprises the following steps: (i) transfecting the host cells with genes which code for at least one protein/product of interest, a neomycin-phosphotransferase, preferably modified, and the amplifiable selectable marker DHFR, wherein in order to enhance the transcription or expression at least the gene (or genes) of interest is functionally linked to at least one TE element; (ii) cultivating the cells under conditions that enable expression of the different genes; (iii) selecting these co-integrated genes by cultivating the cells in the presence of a selecting agent such as e.g.
  • the transfected cells are cultivated in hypoxanthine/thymidine-free medium, supplemented with at least 200 ⁇ g/mL G418, preferably 400 ⁇ g/mL or even more G418, in the absence of serum and with the addition of increasing concentrations of MTX.
  • the concentration of MTX in the first amplification step is at least 100 nM.
  • the concentration of MTX may, however, also be at least 250 nM and may be increased step by step to 1 ⁇ M. In individual cases concentrations of over 1 ⁇ M may also be used, e.g. 2 ⁇ M.
  • FACS fluorescence-activated cell sorting
  • bacterial ⁇ -galactosidase, cell surface markers or fluorescent proteins may be used (e.g. green fluorescent protein (GFP) and the variants thereof from Aequorea Victoria and Renilla reniformis or other species; red fluorescent proteins and proteins which fluoresce in other colours and their variants from non-bioluminescent organisms such as e.g. Discosoma sp., Anemonia sp., Clavularia sp., Zoanthus sp., Aequorea coerulescens ) for the selection of transformed cells.
  • GFP green fluorescent protein
  • red fluorescent proteins and proteins which fluoresce in other colours and their variants from non-bioluminescent organisms such as e.g. Discosoma sp., Anemonia sp., Clavularia sp., Zoanthus sp., Aequorea coerulescens
  • the term gene expression relates to the transcription and/or translation of a heterologous gene sequence in a host cell.
  • the expression rate can be generally determined, either on the basis of the quantity of corresponding mRNA which is present in the host cell or on the basis of the quantity of gene product produced which is encoded by the gene of interest.
  • the quantity of mRNA produced by transcription of a selected nucleotide sequence can be determined for example by northern blot hybridisation, ribonuclease-RNA-protection, in situ hybridisation of cellular RNA or by PCR methods (e.g. quantitative PCR) (Sambrook et al., 1989; Ausubel et al., 1994).
  • Proteins which are encoded by a selected nucleotide sequence can also be determined by various methods such as, for example, ELISA, protein A HPLC, western blot, radioimmunoassay, immunoprecipitation, detection of the biological activity of the protein, immune staining of the protein followed by FACS analysis or fluorescence microscopy, direct detection of a fluorescent protein by FACS analysis or fluorescence microscopy (Sambrook et al., 1989; Ausubel et al., 1994). These methods makes it possible for example to investigate whether the TE element of SEQ ID No. 1 according to the invention, or any part, fragment or region thereof or the derivatives or combinations thereof, lead to an increase in the transcription or expression of a gene of interest.
  • transcription or productivity is meant an enhancement of the expression or synthesis of a heterologous sequence introduced into a host cell, for example a gene coding for a therapeutic protein, compared to a control.
  • a cell according to the invention is cultivated by a method described here according to the invention, and if this cell has at least a doubling of the specific productivity.
  • transcription or productivity if the cell according to the invention has at least a tripling of the specific productivity.
  • transcription or productivity if the cell according to the invention has at least a quadrupling of the specific productivity.
  • transcription or productivity if the specific productivity of the cell according to the invention is increased at least five-fold. There is particularly enhanced expression, transcription or productivity if the specific productivity of the cell according to the invention is increased at least six-fold. There is particularly enhanced expression, transcription or productivity if the specific productivity of the cell according to the invention is increased at least seven-fold.
  • Enhanced expression, transcription or productivity can be achieved both by using one of the expression vectors according to the invention and by using one of the methods according to the invention.
  • the corresponding processes may be combined with a FACS-assisted selection of recombinant host cells which contain, as additional selectable marker, one or more fluorescent proteins (e.g. GFP) or a cell surface marker.
  • fluorescent proteins e.g. GFP
  • Other methods of obtaining increased expression, and a combination of different methods may also be used, are based for example on the use of cis-active elements for manipulating the chromatin structure (e.g.
  • LCR, UCOE, EASE, isolators, S/MARs, STAR elements on the use of (artificial) transcription factors, treatment of the cells with natural or synthetic agents for up-regulating endogenous or heterologous gene expression, improving the stability (half-life) of mRNA or the protein, improving the initiation of mRNA translation, increasing the gene dose by the use of episomal plasmids (based on the use of viral sequences as replication origins, e.g. SV40, polyoma, adenovirus, EBV or BPV), the use of amplification-promoting sequences (Hemann et al., 1994) or in vitro amplification systems based on DNA concatemers (Monaco et al., 1996).
  • episomal plasmids based on the use of viral sequences as replication origins, e.g. SV40, polyoma, adenovirus, EBV or BPV
  • amplification-promoting sequences Hemann et al
  • the present invention thus also relates to processes for obtaining and selecting recombinant mammalian cells which express at least one heterologous gene of interest and are characterised in that (i) recombinant mammalian cells are transfected with an expression vector according to the invention and the gene for an amplifiable selectable marker gene; (ii) the mammalian cells are cultivated under conditions which allow expression of the gene or genes of interest, the modified neomycin phosphotransferase gene and the gene which codes for a fluorescent protein; (iii) the mammalian cells are cultivated in the presence of at least one selecting agent which acts selectively on the growth of mammalian cells and gives preference to the growth of those cells which express the neomycin phosphotransferase gene; (iv) the mammalian cells which exhibit high expression of the fluorescent protein are sorted out by flow-cytometric analysis; (v) the sorted cells are cultivated under conditions under which the amplifiable selectable marker gene is expressed
  • Also preferred according to the invention is a process in which production cells are replicated and used to prepare the coding gene product of interest.
  • the selected high producing cells are preferably cultivated in a serum-free culture medium and preferably in suspension culture under conditions which allow expression of the gene of interest.
  • the protein/product of interest is preferably obtained from the cell culture medium as a secreted gene product. If the protein is expressed without a secretion signal, however, the gene product may also be isolated from cell lysates.
  • conventional purification procedures are carried out. First of all, cells and cell debris are removed from the culture medium or lysate.
  • the desired gene product can then be freed from contaminating soluble proteins, polypeptides and nucleic acids, e.g. by fractionation on immunoaffinity and ion exchange columns, ethanol precipitation, reversed phase HPLC or chromatography on Sephadex, silica or cation exchange resins such as DEAE.
  • Methods which result in the purification of a heterologous protein expressed by recombinant host cells are known to the skilled man and described in the literature, e.g. by Harris et al., 1995 and Scopes 1988.
  • the present invention relates to a nucleic acid which contains TE-13 (SEQ ID No. 15) contains or a fragment of TE-13 (SEQ ID No. 15) or the complementary nucleotide sequences thereof or a derivative of TE-13 (SEQ ID No. 15) or a fragment thereof or the complementary nucleotide sequences thereof, and which on chromosomal integration leads to an increase in the transcription or expression of a gene of interest in an expression system.
  • the present invention relates to a nucleic acid which contains TE-13 (SEQ ID No. 15) or a fragment of TE-13 (SEQ ID No. 15) or the complementary nucleotide sequences thereof or a derivative of TE-13 (SEQ ID No. 15) or a fragment thereof or the complementary nucleotide sequences thereof, and which on chromosomal integration leads to an increase in the transcription or expression of a gene of interest in an expression system, with the proviso that the fragment comprises at least one sequence region from the nucleic acid region between lbp and 1578 bp (in relation to SEQ ID No. 01).
  • the present invention relates to a nucleic acid which contains TE-13 (SEQ ID No. 15) or a fragment of TE-13 (SEQ ID No. 15) or the complementary nucleotide sequences thereof or a derivative of TE-13 (SEQ ID No. 15) or a fragment thereof or the complementary nucleotide sequences thereof, and which on chromosomal integration leads to an increase in the transcription or expression of a gene of interest in an expression system, with the proviso that the fragment comprises at least one sequence region of TE-09 (SEQ ID No. 11) or TE-08 (SEQ ID No. 11) or TE-13 (SEQ ID No. 15).
  • a sequence region is meant a nucleic acid region of at least 10 bp, 15 bp, 20 bp, 50 bp, 100 bp.
  • the enhanced expression of the gene of interest can be measured for example by measuring the product titre by ELISA.
  • the invention relates to a nucleic acid which contains TE-08 (SEQ ID No. 10) or a fragment of TE-08 (SEQ ID No. 10) or the complementary nucleotide sequences thereof or a derivative of TE-08 (SEQ ID No. 10) or a fragment thereof or the complementary nucleotide sequences thereof, and which on chromosomal integration leads to an increase in the transcription or expression of a gene of interest in an expression system.
  • the proviso is that the fragment must also include at least one sequence region from the nucleic acid region between 1 bp and 1578 bp (in relation to SEQ ID No. 01).
  • the fragment also includes at least one sequence region of TE-09 (SEQ ID No. 11) or TE-08 (SEQ ID No. 11) or TE-13 (SEQ ID No. 15).
  • sequence region a nucleic acid region of at least 10 bp, 15 bp, 20 bp, 50 bp, 100 bp.
  • the invention relates to a nucleic acid which contains SEQ ID No. 1 or a fragment of SEQ ID No. 1 or the complementary nucleotide sequences thereof or a derivative of SEQ ID No. 1 or a fragment thereof or the complementary nucleotide sequences thereof, which on chromosomal integration leads to an increase in the transcription or expression of a gene of interest in an expression system.
  • the fragment also comprises at least one sequence region from the nucleic acid region between 1 bp and 1578 bp (in relation to SEQ ID No. 01).
  • the fragment also comprises at least one sequence region of TE-09 (SEQ ID No. 11) or TE-08 (SEQ ID No. 11) or TE-13 (SEQ ID No. 15).
  • a sequence region is meant a nucleic acid region of at least 10 bp, 15 bp, 20 bp, 50 bp, 100 bp.
  • TE element a nucleic acid
  • the increase in the expression of the gene of interest can be measured for example by measuring the product titre by ELISA.
  • the present invention particularly relates to a nucleic acid according to the invention, which hybridises under stringent conditions
  • nucleic acid sequence which has at least about 70% sequence identity, preferably at least about 80% sequence identity, preferably at least about 85% sequence identity, most preferably at least about 90% sequence identity and most preferably at least about 95% sequence identity with (a) or (b),
  • the present invention relates to the 5′ fragment of the TE element TE-00 (SEQ ID No. 2). This corresponds to the part of SEQ ID No. 1 between 1 bp and 1578 bp or the complementary nucleotide sequence thereof.
  • the present invention particularly relates to a nucleic acid or a transcription-enhancing or expression-enhancing nucleic acid element (TE element), which contains TE-13 (SEQ ID No. 15) or TE-08 (SEQ ID No. 10) or a derivative thereof or the complementary nucleotide sequences thereof, which on chromosomal integration leads to an increase in the transcription or expression of a gene of interest.
  • TE element transcription-enhancing or expression-enhancing nucleic acid element
  • the present invention preferably relates to an isolated nucleic acid or an isolated nucleic acid molecule or an isolated nucleic acid sequence or an isolated transcription-enhancing nucleic acid element or an isolated TE element.
  • the present invention particularly relates to an isolated nucleic acid which contains TE-08 (SEQ ID No. 10) or the complementary nucleotide sequence thereof and which on chromosomal integration leads to an increase in the transcription or expression of a gene of interest in an expression system.
  • the nucleic acid or the transcription-enhancing nucleic acid element or the isolated nucleic acid contains a derivative of a TE element or of SEQ ID No. 1, which has at least about 70% sequence identity, preferably at least about 80% sequence identity, most preferably at least about 90% sequence identity and most preferably at least about 95% sequence identity with the corresponding part of the TE element sequence or the complementary sequence thereof, particularly with the sequence region between nucleotide position 1 bp and 1578 bp in relation to SEQ ID No. 1, corresponding to the sequence region 5′ of the TE-00 sequence, and particularly preferably with TE-13 (SEQ ID No. 15) or TE-08 (SEQ ID No. 10).
  • the nucleic acid or the transcription-enhancing nucleic acid element or the isolated nucleic acid contains a derivative of a TE-08 nucleic acid (SEQ ID No. 10) or preferably of a TE-13 nucleic acid (SEQ ID No. 15), which has at least about 70% sequence identity, preferably at least about 80% sequence identity, most preferably at least about 90% sequence identity and most preferably at least about 95% sequence identity with the corresponding part of the TE element sequence or the complementary sequence thereof.
  • the invention relates to a nucleic acid or a transcription-enhancing nucleic acid element or an isolated nucleic acid or a derivative of a TE element, which hybridise(s) with the sequence of a TE element or with the complementary sequence of a TE element, particularly with the sequence region between nucleotide position 1 bp and 1578 bp in relation to SEQ ID No. 1, corresponding to sequence region 5′ of the TE-00 sequence (SEQ ID No. 2), or hybridises particularly with the TE-08 element (SEQ ID No. 10).
  • the hybridisation is carried out under stringent hybridisations and washing conditions.
  • the nucleic acid according to the invention or the transcription-enhancing element (TE element) is selected from among: TE-00 (SEQ ID No. 2), TE-01 (SEQ ID No. 3), TE-02 (SEQ ID No. 4), TE-03 (SEQ ID No. 5), TE-04 (SEQ ID No. 6), TE-06 (SEQ ID No. 8), TE-07 (SEQ ID No. 9), TE-08 (SEQ ID No. 10), TE-10 (SEQ ID No. 12), TE-11 (SEQ ID No. 13), TE-12 (SEQ ID No. 14), TE-13 (SEQ ID No. 15), TE-14 (SEQ ID No. 16), TE-15 (SEQ ID No. 17), TE-16 (SEQ ID No. 18), TE-17 (SEQ ID No. 19), TE-18 (SEQ ID No. 20) and TE-21 (SEQ ID No. 21).
  • TE-00 SEQ ID No. 2
  • TE-01 SEQ ID No. 3
  • nucleic acid or the transcription-enhancing element is characterised in that the nucleic acid is TE-00 (SEQ ID No. 2), TE-06 (SEQ ID No. 8), TE-10 (SEQ ID No. 12), TE-11 (SEQ ID No. 13) or TE-12 (SEQ ID No. 14), preferably TE-06 (SEQ ID No. 8).
  • nucleic acid or the transcription-enhancing element is characterised in that the nucleic acid is TE-01 (SEQ ID No. 3), TE-02 (SEQ ID No. 4), TE-03 (SEQ ID No. 5),TE-07 (SEQ ID No. 9), TE-08 (SEQ ID No. 10).
  • nucleic acid or the transcription-enhancing element is TE-08 (SEQ ID No. 10).
  • the nucleic acid or the transcription-enhancing element is characterised in that it is a fragment or derivative of TE-01 (SEQ ID No. 3) is, preferably TE-13 (SEQ ID No. 15), TE-14 (SEQ ID No. 16), TE-15 (SEQ ID No. 17), TE-16 (SEQ ID No. 18), TE-17 (SEQ ID No. 19), TE-18 (SEQ ID No. 20).
  • nucleic acid according to the invention is TE-13 (SEQ ID No. 15).
  • nucleic acid according to the invention or the fragment or the derivative is an isolated nucleic acid.
  • the present invention also relates to a nucleic acid according to the invention according to one of claims 1 to 12 , characterised in that a nucleic acid containing TE-13 (SEQ ID No. 15) or TE-08 (SEQ ID No. 10) or TE-07 (SEQ ID No. 9) or TE-06 (SEQ ID No. 8) or a fragment of these sequences or the complementary nucleotide sequences thereof or a derivative of these sequences or a derivative of fragments of these sequences, preferably TE-13 (SEQ ID No. 15) or TE-08 (SEQ ID No. 10) or a fragment of these sequences or a derivative of these sequences or a derivative of fragments of these sequences or the complementary nucleotide sequences thereof, is linked to a heterologous sequence.
  • the nucleic acid linked to a heterologous gene sequence may in a preferred embodiment be an expression vector, for example plasmids, bacteriophages, phagemids, cosmids, viral vectors or particularly a targeting vector.
  • the nucleic acid linked to a heterologous gene sequence may however also be any other synthetic nucleic acid molecule such as e.g. synthetic, artificial or mini-chromosomes.
  • the present invention further relates to a eukaryotic expression vector, characterised in that this expression vector contains one or more nucleic acids according to the invention or one or more transcription-enhancing elements (TE element) according to the invention.
  • this expression vector contains one or more nucleic acids according to the invention or one or more transcription-enhancing elements (TE element) according to the invention.
  • the eukaryotic expression vector is characterised in that it contains a promoter and/or a heterologous gene of interest and/or a selectable marker and/or optionally an enhancer.
  • the present invention further relates to a eukaryotic expression vector characterised in that this expression vector contains one or more nucleic acids according to the invention or one or more transcription-enhancing elements (TE element) and a promoter and/or a heterologous gene of interest and/or a selectable marker and/or optionally an enhancer according to the invention.
  • TE element transcription-enhancing elements
  • the eukaryotic expression vector is characterised in that it is a targeting vector for the deliberate integration of the gene of interest.
  • the eukaryotic expression vector is characterised in that the promoter is a heterologous promoter such as the early promoter of human cytomegaly virus (CMV-promoter), SV40 early promoter, adenovirus major late promoter, mouse metallothionein-I promoter, the long terminal repeat region of Rous Sarcoma Virus, actin-, immunoglobulin or heat-shock-promotor(s), preferably CMV-promoter.
  • CMV-promoter human cytomegaly virus
  • SV40 early promoter SV40 early promoter
  • adenovirus major late promoter adenovirus major late promoter
  • mouse metallothionein-I promoter mouse metallothionein-I promoter
  • the long terminal repeat region of Rous Sarcoma Virus actin-
  • immunoglobulin or heat-shock-promotor(s) preferably CMV-promoter.
  • the eukaryotic expression vector is characterised in that the promoter is a heterologous promoter, preferably ubiquitin/S27a-promoter, most preferably hamster ubiquitin/S27a-promoter.
  • the eukaryotic expression vector is characterised in that it contains a combination of several identical or different nucleic acids or TE-elements according to the invention in any orientation to one another, wherein one or more nucleic acids or TE-elements are positioned in front of (i.e. 5′ of) and/or one or more nucleic acids or TE-elements are positioned (i.e. 3′ of) the gene of interest.
  • the eukaryotic expression vector is characterised in that the combined nucleic acids or TE elements are TE-06 (SEQ ID No. 8) and particularly preferably TE-08 (SEQ ID No. 10).
  • Preferred combinations of nucleic acids or TE elements are a TE-06 element (SEQ ID No. 8) in front of (i.e. 5′ of) and a TE-06 element (SEQ ID No. 8) behind (i.e. 3′ of) the gene of interest and three TE-06-elements (SEQ ID No. 8) behind (i.e. 3′ of) the gene of interest (cf also FIG. 13 ).
  • the eukaryotic expression vector is characterised in that one or more TE-08-nucleic acid(s) or element(s) (SEQ ID No. 10) are positioned in front of (i.e. 5′ of) and one or more behind (i.e. 3′ of) the gene of interest, preferably one TE-08 element (SEQ ID No. 10) in front and one behind (cf also FIG. 13 ).
  • TE-08-nucleic acid(s) or element(s) SEQ ID No. 10
  • Other preferred combinations of TE-nucleic acids or elements are 2 TE-08 nucleic acids/elements (SEQ ID No. 10) before and/or after the gene of interest.
  • the eukaryotic expression vector is characterised in that a plurality of TE-06-nucleic acids or elements (SEQ ID No. 8) are positioned behind (3′ of) the gene of interest, preferably 3 (cf FIG. 13 ).
  • the eukaryotic expression vector is characterised in that a combination of one or more TE-08-nucleic acid(s) or element(s) (SEQ ID No. 10) with one or more TE-06 nucleic acids or element(s) (SEQ ID No. 8) are positioned in front of (i.e. 5′ of) and/or behind (i.e. 3′ of) the gene of interest; the preferred combination is a combination of a TE-08 nucleic acid or element (SEQ ID No. 10) followed by a TE-06-nucleic acid or element (SEQ ID No. 8) in front of (i.e. 5′ of) the gene of interest. (Cf also FIG. 13 ).
  • the eukaryotic expression vector is characterised in that it additionally contains an integrase.
  • the host cells are co-transfected with at least two eukaryotic expression vectors, while at least one of the two vectors contains at least one gene which codes for at least one protein of interest and the other vector contains one or more nucleic acid according to the inventions in any combination, position and orientation, and optionally also codes for at least one gene of interest, and these nucleic acids according to the invention impart their transcription- or expression-enhancing effect to the genes of interest which are located on the other co-transfected vector, by co-integration with the other vector.
  • the eukaryotic expression vector is characterised in that the selectable marker is DHFR or Neo, for example Neo F240I.
  • the invention further relates to a method of preparing a eukaryotic expression vector, characterised by the integration of a nucleic acid according to the invention in an expression vector.
  • the invention further relates to a eukaryotic host cell characterised in that it contains a eukaryotic expression vector according to the invention.
  • the eukaryotic host cell is characterised in that it is a high producer, i.e. it has a higher specific productivity than a comparable eukaryotic host cell without a TE element or nucleic acid according to the invention, this host cell having an expression level which is increased two-fold, three-fold, four-fold, five-fold, six-fold, seven-fold or ten-fold or one which is increased more than two-fold, more than three-fold, more than four-fold, more than five-fold, more than seven-fold or more than ten-fold, preferably up to five-fold or more than three-fold.
  • the eukaryotic host cell is characterised in that the expression vector is stably integrated in the genome.
  • the eukaryotic host cell is a hamster or mouse cell such as for example a CHO, NS0, Sp2/0-Ag14, BHK21, BHK TK ⁇ , HaK, 2254-62.2 (BHK-21-derivative), CHO-K1, CHO-DUKX ( ⁇ CHO duk ⁇ , CHO/dhfr ⁇ ), CHO-DUKX B1, CHO-DG44, CHO Pro-5, V79, B14AF28-G3, CHL cell, preferably a CHO cell and particularly preferably a CHO-DG44 cell.
  • the eukaryotic host cell is a mammalian cell, including but not restricted to human, mouse, rat, monkey or rodent cell lines.
  • the host cell is a eukaryotic cell including but not restricted to yeast, insect, bird and plant cells.
  • the eukaryotic host cell is characterised in that it additionally contains an anti-apoptosis gene such as BCL-xL, BCL-2, BCL-w, BFL-1, A1, MCL-1, BOO, BRAG-1, NR-13, CDN-1, CDN-2, CDN-3, BHRF-1, LMW5-HL or CED-9, preferably Bcl-xL or BCL-2, most preferably BCL-xL.
  • an anti-apoptosis gene such as BCL-xL, BCL-2, BCL-w, BFL-1, A1, MCL-1, BOO, BRAG-1, NR-13, CDN-1, CDN-2, CDN-3, BHRF-1, LMW5-HL or CED-9, preferably Bcl-xL or BCL-2, most preferably BCL-xL.
  • the present invention further relates to a method of developing a high-producing stably transfected eukaryotic host cell line, characterised by the following steps:
  • the present invention further relates to a method of developing a high-producing stably transfected eukaryotic host cell line, characterised by the following steps:
  • the method is characterised by at least one additional amplification step.
  • the present invention also relates to a method of preparing and selecting recombinant mammalian cells, characterised by the following steps:
  • this method is characterised in that the transfected cells are cultivated in hypoxanthine/thymidine-free medium, supplemented with at least 200 ⁇ g/mL G418, preferably 400 ⁇ g/mL or more G418, in the absence of serum and with the addition of increasing concentrations of MTX.
  • this method is characterised in that the concentration of MTX in the first amplification step is at least 100 nM or at least 250 nM and is increased stepwise to 1 ⁇ M or above. In individual cases, the MTX concentration may be 2 ⁇ M.
  • the method is characterised by an additional cloning step.
  • the host cell is a rodent/hamster cell such as for example a CHO, NS0, Sp2/0-Ag14, BHK21, BHK TK ⁇ , HaK, 2254-62.2 (BHK-21-derivative), CHO-K1, CHO-DUKX ( ⁇ CHO duk ⁇ , CHO/dhfr ⁇ ), CHO-DUKX B1, CHO-DG44, CHO Pro-5, V79, B14AF28-G3, CHL cell, preferably a CHO cell and particularly preferably a CHO-DG44 cell.
  • rodent/hamster cell such as for example a CHO, NS0, Sp2/0-Ag14, BHK21, BHK TK ⁇ , HaK, 2254-62.2 (BHK-21-derivative), CHO-K1, CHO-DUKX ( ⁇ CHO duk ⁇ , CHO/dhfr ⁇ ), CHO-DUKX B1, CHO-DG44, CHO Pro-5, V79, B
  • the expression vector contains a selectable marker such as DHFR or NPT, for example NPT F240I or NPT D227G.
  • the proportion of high producers is increased up to two-fold, three-fold, four-fold, five-fold, six-fold, seven-fold or ten-fold or more than two-fold, more than three-fold, more than four-fold, more than five-fold, more than seven-fold or more than ten-fold, preferably up to five-fold or more than three-fold.
  • the present invention further relates to a method of preparing a biopharmaceutical product, characterised by the following steps:
  • the present invention further relates to a method of developing a high-producing stably transfected eukaryotic host cell line, characterised by the following steps:
  • the method according to the invention is characterised by at least one additional amplification step.
  • the present invention further relates to the use of a nucleic acid according to the invention or of a transcription-enhancing element (TE element) according to the invention in a eukaryotic expression vector, for increasing the transcription or expression of a gene of interest in an expression system in a eukaryotic host cell or for preparing a biopharmaceutical product.
  • a transcription-enhancing element TE element
  • the present invention also relates to the use of a nucleic acid according to the invention or of a transcription-enhancing element (TE element) according to the invention for producing transgenic animals or plants.
  • TE element transcription-enhancing element
  • the present invention further relates to the use of a nucleic acid according to the invention or of a transcription-enhancing element (TE element) according to the invention in gene therapy.
  • TE element transcription-enhancing element
  • the present invention particularly relates to the use of a nucleic acid according to the invention or of a transcription-enhancing element (TE element) according to the invention as a medicament or in a pharmaceutical composition.
  • a transcription-enhancing element TE element
  • the present invention further relates to a kit consisting of a nucleic acid according to the invention or (a) TE element(s) according to the invention, optionally expression vector(s), optionally host cell(s) and optionally transfection reagent(s).
  • the present invention relates to a nucleic acid, particularly an isolated nucleic acid, more precisely a transcription-enhancing or expression-enhancing nucleic acid element (TE element) with SEQ ID No. 1 or a fragment or a derivative thereof, which on chromosomal integration leads to an increase in the transcription or expression of a gene of interest, with the exclusion of the TE element TE-00 (SEQ ID No. 2).
  • TE element transcription-enhancing or expression-enhancing nucleic acid element
  • the present invention relates to a nucleic acid according to the invention with the exclusion of the TE elements TE-00 (SEQ ID No. 2), TE-04 (SEQ ID No. 6) and TE-06 (SEQ ID No. 8).
  • the present invention relates to a nucleic acid according to the invention with the exclusion of the TE elements TE-00 (SEQ ID No. 2), TE-04 (SEQ ID No. 6), TE-05 (SEQ ID No. 7) and TE-06 (SEQ ID No. 8).
  • the increase in the expression of the gene of interest can be measured for example by measuring the product titre using ELISA.
  • a TE element, fragment or derivative according to the invention which is over 160 bp long, preferably over 170 bp long.
  • the TE element fragment is between 160 bp and 1.2 kb or between 170 bp and 1 kb, preferably over 200 bp and between 200 bp and 1 kb long.
  • the TE element fragment is in the part of SEQ ID No. 1 between 1 bp and 1578 bp (this corresponds to a fragment 5′ of the element TE-00 (SEQ ID No. 2) and is over 113 bp long or over 132 bp and preferably over 160 bp or over 170 bp long.
  • the TE element fragment is between 113 bp and 1.2 kb or between 132 bp and 1.2 kb or between 160 bp and 1.2 kb, preferably over 200 bp and between 200 bp and 1 kb long.
  • the TE element fragment is present without any adjacent sequences.
  • the fragment is not part of a larger sequence or a sequence region, for example no other sequences are attached in front of (5′) or behind (3′) it.
  • the present invention relates to a nucleic acid according to the invention which does not contain any CpG islands.
  • the collection of data on the product titre and specific productivity showed that on average almost all the cell pools with TE elements which were fragments or derivatives of SEQ ID No. 1 expressed more genes of interest than cell pools without a TE element.
  • the TE-elements 01 (SEQ ID No. 3), 02 (SEQ ID No. 4) and 08 (SEQ ID No. 10) yielded the highest productivity in two independent transfection series in which a different selectable marker (NPT or DHFR) was used in each case. They are capable of increasing the productivity by a factor of 4-7.
  • the TE elements 01 (SEQ ID No. 3) and 02 (SEQ ID No. 4) at 3 kb and 2.5 kb are very large for additionally attaching to an expression vector.
  • the TE element 08 (SEQ ID No. 10), which is only 1 kb in size, which is capable of increasing expression by a factor of 5-6.
  • the TE element 06 (SEQ ID No. 8) in two transfection series yielded a tripling of the specific productivity with a size of only 381 bp. It is highly advantageous to leave the expression vectors as small as possible, as smaller vectors are generally more stable and are easier to handle both during cloning and during transfection. Therefore, the TE-elements 06 (SEQ ID No. 8) and 08 (SEQ ID No. 10) are particularly interesting for use as transcription-promoting elements.
  • the following Examples show that the cell pools which contained the TE element 03 (SEQ ID No. 5), 04 (SEQ ID No. 6) or 07 (SEQ ID No. 9) showed an approximately 3- to 3.5-fold expression of the gene of interest and cell pools with the TE elements 10 (SEQ ID No. 12), 11 (SEQ ID No. 13) or 12 (SEQ ID No. 14) showed an approximately doubled expression of the gene of interest
  • the TE element 08 in conjunction with NPT F240I as selectable marker, at factor 5, demonstrated the greatest increase in the specific productivity of the gene of interest compared with the control pools with no TE element.
  • the TE element 08 shows an even better increase of about factor 6.0.
  • TE element 02 SEQ ID No. 4 in the test series with DHFR as selectable marker shows an increase in the productivity of the gene of interest by a factor 6.8.
  • FACS fluorescence-activated cell sorter
  • GFP green fluorescent protein
  • IgG Immunoglobulin G
  • IRES internal ribosomal entry site
  • mAb monoclonal antibody
  • MCP-1 monocyte chemoattractant protein-1
  • NPT neomycin-phosphotransferase
  • SEAP secreted alkaline phosphatase
  • the cells CHO-DG44/dhfr ⁇ / ⁇ (Urlaub et al., 1983) were permanently cultivated as suspension cells in serum-free CHO-S-SFMII medium supplemented with hypoxanthine and thymidine (HT) (Invitrogen GmbH, Düsseldorf, DE) in cell culture flasks at 37° C. in a damp atmosphere and 5% CO 2 .
  • the cell counts and viability were determined with a Coulter Counter Z2 (Beckmann Coulter) or with a Cedex (Innovatis) and the cells were then seeded in a concentration of 1-3 ⁇ 10 5 /mL and run every 2-3 days.
  • Lipofectamine Plus Reagent (Invitrogen GmbH) was used for the transfection of CHO-DG44.
  • a total of 1.0-1.3 ⁇ g of plasmid-DNA, 4 ⁇ L of lipofectamine and 6 ⁇ L of Plus reagent were mixed together according to the manufacturer's instructions and added in a volume of 200 ⁇ L to 6 ⁇ 10 5 cells in 0.8 mL of HT-supplemented CHO-S-SFMII medium. After three hours' incubation at 37° C. in a cell incubator 2 mL of HT-supplemented CHO-S-SFMII medium was added. After a is cultivation time of 48 hours, the transfection mixtures were either harvested (transient transfection) or subjected to selection.
  • the cells were transferred 2 days after transfection into HT-supplemented CHO-S-SFMII medium with 400 ⁇ g/mL of G418 (Invitrogen).
  • the cells were transferred 2 days after transfection into HT-free CHO-S-SFMII medium.
  • the cells were transferred 2 days after transfection into CHO-S-SFMII medium without the addition of hypoxanthine and thymidine and also G418 (Invitrogen) was added to the medium in a concentration of 400 ⁇ g/mL.
  • a DHFR-based gene amplification of the integrated heterologous genes can be obtained by the addition of the selecting agent MTX (Sigma, Deisenhofen, DE) in a concentration of 5-2000 nM to an HT-free CHO-S-SFMII medium.
  • MTX Sigma, Deisenhofen, DE
  • eukaryotic expression vectors were used which are based on the pAD-CMV vector (Werner et al., 1998) and mediate the expression of a heterologous gene by the combination of CMV enhancer/hamster ubiquitin/S27a promoter (WO 97/15664) or CMV enhancer/CMV promoter.
  • the base vector pBID contains the dhfr-minigene which acts as an amplifiable selectable marker (cf e.g. EP-0-393-438)
  • the vector pBING the dhfr-minigene has been replaced by a modified NPT gene. This is the NPT variant D227G (Asp227Gly).
  • the cloning of pBING with the NPT variant D227G and the IRES-GFP gene region was carried out as described in (WO2004/050884).
  • the base plasmid pTE4 contains the NPT variant F2401 (Phe240IIe) as selectable marker and is a derivative of the plasmid pBING.
  • the GFP was also replaced by the red fluorescent protein DsRed2 from the vector pDsRed2 (Clontech, Palo Alto, Calif., USA).
  • the base plasmid pTE5 contains DHFR as selectable marker and is a derivative of the vector pBJDG (WO2004/050884), in which again the GFP has been replaced by the red fluorescent protein DsRed2 from the vector pDsRed2 (Clontech, Palo Alto, Calif., USA).
  • the heavy chain was cloned as a 1.4 kb SalI/SpeI fragment into the plasmid pBID digested with XbaI and SalI, to obtain the plasmid pBID-HC ( FIG. 1A ).
  • the light chain on the other hand was cloned as a 0.7 kb BamHI/HindIII fragment into the cutting sites of the plasmid pBING, producing the plasmid pBING-LC ( FIG. 1A ).
  • Human MCP-1 cDNA (Yoshimura et al., 1989) was cloned into the corresponding cutting sites of the vector pTE4 or pTE5 as a 0.3 kb HindIII/EcoRI fragment, resulting in the plasmids pTE4/MCP-1 and pTE5/MCP-1 ( FIG. 1B and FIG. 2 respectively).
  • the flow-cytometric analyses were carried out with a BD FACScalibur (BD Bioscience).
  • the FACS is fitted with a helium-argon laser with an excitation wavelength of 488 nm.
  • the fluorescence intensity is absorbed at a wavelength suited to the particular fluorescence protein and processed by means of the attached software Cell Quest Pro.
  • the MCP-1 titres in supernatants of stably or transiently transfected CHO-DG44 cells were quantified by ELISA using the OptElA Human MCP-1 Set kit in accordance with the manufacturer's instructions (BD Biosciences Pharmingen, Heidelberg, Del.).
  • the IgG1 mAb in the supernatants from stably transfected CHO-DG44 cells was quantified by ELISA according to standard procedures (Current Protocols in Molecular Biology, Ausubel et al., 1994, updated), using on the one hand a goat anti human IgG Fc fragment (Dianova, Hamburg, Del.) and on the other hand an AP-conjugated goat anti human kappa light chain antibody (Sigma). Purified IgG1 antibody was used as the standard.
  • SEAP titre in culture supernatants from transiently transfected CHO-DG44 cells was quantified using the SEAP reporter gene assays in accordance with the manufacturer's instructions (Roche Diagnostics GmbH, Mannheim, Del.).
  • nucleotides 62 to 85 corresponds to nucleotides 62 to 85 (complementary sequence) of SEQ ID No. 5 from WO 97/15664).
  • a second PCR was carried out with a second primer combination, consisting of an inner adapter primer and an inner (“nested”) hamster-specific primer (primer Ub21: 5′-GGGTTTCTCTGTGTAATAGCCATG-3 (SEQ ID No. 40); corresponds to nucleotides 16 to 39 (complementary sequence) of SEQ ID No. 5 from WO97/15664).
  • the amplified material obtained was subcloned into the pCR2.1 TOPO vector (Invitrogen) and sequenced. It contained another 1239 bp of a new, hitherto unknown sequence upstream from the hamster Ub/S27a-gene.
  • sequence information from the overlapping PCR fragments was used to amplify a cohesive sequence region from the genomic DNA of CHO-DG44 by PCR, which comprised all the partial fragments hitherto isolated and extended 383 bp into the 5′ sequence region of SEQ ID No. 5 from WO 97/15664, using the primers Ub34 (5′-CTAAGAGTACTTGCCATGAGAGCCTGAA-3′ (SEQ ID No. 43); located at the outermost 5 end of the newly isolated 1239 bp sequence; only partially present in SEQ ID No. 1 (nucleotides 1 to 14)) and
  • Ub35 (5′-CATTGATACACCACCAAAGAACTTG-3′ (SEQ ID No. 44); corresponds to nucleotides 1941 to 1965 (complementary sequence) of SEQ ID No. 1).
  • the resulting 2 kb DNA fragment was ligated with the 5′ UTR region of sequence ID No. 5 described in WO 97/15664 via the endogenous EcoRI cutting site (position 353-358), although this cutting site was eliminated by a filling reaction with Klenow DNA polymerase.
  • a second endogenous EcoRl cutting site in the newly isolated genome region was eliminated in the same way, resulting in the nucleotides 326 to 329 in SEQ ID No. 1 which are additional to the original genome sequence. In all, in this way, 8 additional nucleotides were inserted into SEQ ID No. 1 compared with the endogenous hamster sequence.
  • the resulting 3788 bp DNA fragment from a sequence region located upstream from the hamster Ub/S27a gene was designated TE element A with the sequence ID No. 1 and subcloned into the vector pBluescript SKM (Stratagene, La Jolla, Calif.).
  • the fragment TE-00 (SEQ ID No. 2) was isolated from a subclone of TE-A by SacII-restriction enzyme digestion and cloned into the base vectors pBING-LC ( FIG. 1A ) or pBID-HC ( FIG. 1A ) in both direct and reverse orientation via the SpeI cutting site located 5′ of the promoter/enhancer element.
  • CHO-DG44 cells were co-transfected with the following plasmid combinations or plasmid variants: control plasmids pBING-LC ( FIG. 1A ) and pBID-HC ( FIG. 1A ) without TE element pBING-LC and pBID-HC with a TE element TE-00 integrated upstream from the promoter/enhancer in direct orientation
  • transfection series A four pools were produced, in transfection series B ten pools were produced per variant. Equimolar amounts of the two plasmids were used. In order to arrive at the same total number of molecules, the total amount of DNA used in series A in the control mixtures was 1 ⁇ g, while in the mixtures containing TE element the amount was 1.3 ⁇ g. This difference resulted from the different plasmid sizes, as the plasmids with TE element were larger than the control plasmids by a factor of 1.3.
  • the IgG1 titre and the specific productivity of the pools were determined over a period of six to eight passages (passaging rhythm 2-2-3 days).
  • the cell pools containing the TE element 00 on average expressed more than the cell pools without TE element ( FIG. 9 , series A and B).
  • a doubling of the pool productivity could be demonstrated as a result of the presence of the TE element, while the orientation in which the element was cloned in the expression plasmid was of no relevance.
  • the effect of the TE elements TE-01 to TE-12 on the expression of the secreted MCP-1 was investigated in three stable transfection series (Series C, D and E) of CHO-DG44 cells compared with expression without the TE element. In all three series, 6 pools were produced per plasmid variant.
  • a mock-transfected pool was also run, i.e. treated in the same way but without the addition of DNA to the transfection mixture.
  • the selection of stably transfected cells was carried out two days after transfection, with HT-supplemented CHO-S-SFMII+G418 (400 ⁇ g/mL) in Series C and D and with HT-free CHO-S-SFMII in Series E.
  • FIG. 8 shows the relative percentage fluorescence of the living transfected cells from Series C. Compared with the control pools, the pools which contained the TE elements TE-01, TE-02 or TE-08, contained about 3 to 3.5 times more dsRed2-expressing cells and pools with the element TE-06 contained approximately twice as many dsRed2-expressing cells. In pools with the fragments TE-05 and TE-09, on the other hand, there was no apparent increase in the proportion of dsRed2-expressing cells compared with the control. In addition, the MCP-1 product titre and the specific productivity were also raised over a period of 6 passages (passaging rhythm 2-2-3 days). FIG.
  • FIG. 9 shows a relative specific MCP-1 productivities.
  • the element TE-08 in conjunction with NPT-F240I as selectable marker with factor 5.3 showed the greatest increase in the specific MCP-1 productivity compared with the control pools without TE element ( FIG. 9 ).
  • DHFR as selectable marker, a 6-fold increase was achieved with this variant ( FIG. 10 ).
  • the TE elements 01, 02 and 03 resulted in a 4 to 4.5-fold increase in productivity in the NPT-selected pools ( FIG. 9 ) and a 2.6 to 6.8-fold increase in productivity in the DHFR-selected pools ( FIG. 10 ) compared with the control pools.
  • the TE element 06 which is only 300 bp long was also able to increase productivity in all the series by a factor 2.5 to 3.2 ( FIGS. 9 and 10 ).
  • the increases achieved with fragments TE-04 and TE-07 were also of this order of magnitude ( FIG. 10 ).
  • FIG. 11 shows the average from the 6 parallel pools.
  • the effect of the TE elements TE-13 to TE-18 on the expression of the secreted MCP-1 was investigated in a stable transfection series (Series F) of CHO-DG44 cells by comparison with expression without the TE element.
  • Four pools were produced per plasmid variant.
  • the base plasmid was pTE4/MCP-1 in all the series ( FIG. 1B ; Selectable Marker NPT—Neomycin-phosphotransferase F240I).
  • telomere length varies between 6.7 kb and 8.2 kb.
  • a mock-transfected pool was run at the same time in each transfection series, i.e. treated the same, but without the addition of DNA in the transfection mixture. The selection of stably transfected cells took place two days after transfection, using HT-supplemented CHO-S-SFMII +G418 (400 ⁇ g/mL).
  • FIG. 14 shows the relative specific MCP-1 productivities. Each of the elements leads to an increase in the average MCP-1 expression. The greatest increase (15-fold) was obtained using element 13, which even exceeds the 10-fold increase produced by element 08.
  • the effect of the TE elements TE-06 and TE-08 in various combinations and at various positions in the expression plasmid on the expression of the secreted MCP-1 was investigated in two stable transfection series (Series G and H) of CHO-DG44 cells compared with expression without the TE element. In both series, 6 pools were produced per plasmid variant.
  • Series H the elements TE-06 and TE-21 or TE-08 are used in front of the enhancer/promoter element (E/P) and additionally after the termination signal (T) ( FIG. 13 ).
  • E/P enhancer/promoter element
  • T termination signal
  • the plasmid size varied between 6.7 kb and 10.2 kb.
  • a mock-transfected pool was run at the same time as each transfection series, i.e. treated the same but without the addition of DNA in the transfection mixture.
  • the selection of stably transfected cells took place two days after transfection, with HT-supplemented CHO-S-SFMII+G418 (300 ⁇ g/mL).
  • FIG. 15 shows the relative specific MCP-1 productivities of Series G. All the elements lead to an increase in the average MCP-1 expression. The greatest increase (4-fold) was produced by the element TE-A. The use of the elements TE-06 and TE-21 or TE-08 before and after the expression cassette gave rise to an increase.
  • TE element TE-08 on the expression of two IgG-4 antibodies is investigated in a stable transfection series (Series J) of CHO-DG44 cells by comparison with the expression without the TE element.
  • 24 pools are produced with the base plasmids pBIN-LC2 or.
  • pBID-HC3 and 24 pools are produced with pBIN-LC2/TE08 or.
  • 1.2 ⁇ g of plasmid DNA are used in total in each case.
  • the plasmid size varies between 6.1 kb and 7.5 kb.
  • a mock-transfected pool is run at the same time with each transfection series, i.e. treated the same, but without the addition of DNA to the transfection mixture.
  • the selection of stably transfected cells is carried out two days after transfection, using HT-free CHO-S-SFMII+G418 (400 ⁇ g/mL).
  • IgG-4 product titres and the specific productivity are obtained over a period of 4 passages (passaging rhythm 2-2-3 days).
  • the element 08 leads to an increase in the average expression rate in the expression of IgG4 antibodies.
  • the chance of finding a high producing cell pool can be increased by the presence of the element TE-08.
  • the effect of various TE elements on the expression of the secreted MCP-1 is investigated in a stable transfection series (Series K) of HEK293 freestyle cells by comparison with MCP-1 expression without a TE element.
  • the elements TE-08, TE-13 and TE-A are used in direct orientation upstream from the enhancer/promoter and 7-10 pools are produced per plasmid variant.
  • 1.2 ⁇ g of plasmid DNA are used in total in each case.
  • the plasmid size varies between 6.7 kb and 10.2 kb.
  • a mock-transfected pool is run at the same time as each transfection series, i.e. treated the same but without the addition of DNA to the transfection mixture.
  • the selection of stably transfected cells takes place two days after the transfection with 293 SFM II medium+4 mM glutamin+G418 (100 ⁇ g/ml).
  • MCP-1 product titre and the specific productivity are obtained over a period of 5 to 6 passages (passaging rhythm 2-2-3 days).
  • the effect of the TE element TE-08 on the expression of an enzyme (SEAP) is investigated in a stable transfection series (Series L) of CHO-DG44 cells compared with SEAP expression without the TE element.
  • Six pools are produced per plasmid variant.
  • the base plasmid is pTE-4/SEAP. It is generated by exchanging MCP-1—IRES—DsRed2-expression cassette for SEAP.
  • 1.2 ⁇ g of plasmid DNA are used in total in each case.
  • the plasmid size varies between 6.6 kb and 7.6 kb.
  • a mock-transfected pool is run at the same time as each transfection series, i.e. treated the same but without the addition of DNA to the transfection mixture.
  • the selection of stably transfected cells takes place two days after transfection, with HT-supplemented CHO-S-SFMII+G418 (400 ⁇ g/mL).
  • the relative SEAP expression is determined using the commercially obtainable SEAP assay (Clontech) and obtained over a period of 6 passages (passaging rhythm 2-2-3 days).

Landscapes

  • Health & Medical Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Zoology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biomedical Technology (AREA)
  • Wood Science & Technology (AREA)
  • General Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • General Health & Medical Sciences (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Biochemistry (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Physics & Mathematics (AREA)
  • Medicinal Chemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Toxicology (AREA)
  • Public Health (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US11/762,240 2006-07-26 2007-06-13 Regulatory Nucleic Acid Elements Abandoned US20080124760A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US12/842,468 US9045776B2 (en) 2006-07-26 2010-07-23 Regulatory nucleic acid elements
US14/699,182 US9708626B2 (en) 2006-07-26 2015-04-29 Regulatory nucleic acid elements

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
EP06117862 2006-07-26
EP06117862.0 2006-07-26

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/842,468 Continuation US9045776B2 (en) 2006-07-26 2010-07-23 Regulatory nucleic acid elements

Publications (1)

Publication Number Publication Date
US20080124760A1 true US20080124760A1 (en) 2008-05-29

Family

ID=37441548

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/762,240 Abandoned US20080124760A1 (en) 2006-07-26 2007-06-13 Regulatory Nucleic Acid Elements
US12/842,468 Active 2027-11-28 US9045776B2 (en) 2006-07-26 2010-07-23 Regulatory nucleic acid elements
US14/699,182 Active US9708626B2 (en) 2006-07-26 2015-04-29 Regulatory nucleic acid elements

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/842,468 Active 2027-11-28 US9045776B2 (en) 2006-07-26 2010-07-23 Regulatory nucleic acid elements
US14/699,182 Active US9708626B2 (en) 2006-07-26 2015-04-29 Regulatory nucleic acid elements

Country Status (23)

Country Link
US (3) US20080124760A1 (es)
EP (1) EP2049671B1 (es)
JP (1) JP5270544B2 (es)
KR (1) KR101485853B1 (es)
CN (1) CN101517085B (es)
AR (1) AR061472A1 (es)
AU (1) AU2007278368B2 (es)
BR (1) BRPI0714594A2 (es)
CA (1) CA2658595C (es)
CY (1) CY1113711T1 (es)
DK (1) DK2049671T3 (es)
EA (1) EA016880B1 (es)
ES (1) ES2401654T3 (es)
HK (1) HK1134107A1 (es)
IL (1) IL196676A0 (es)
MX (1) MX2009000781A (es)
NO (1) NO20090057L (es)
NZ (1) NZ575115A (es)
PL (1) PL2049671T3 (es)
PT (1) PT2049671E (es)
SI (1) SI2049671T1 (es)
TW (1) TW200815591A (es)
WO (1) WO2008012142A1 (es)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9085627B2 (en) 2011-04-13 2015-07-21 National Research Council Of Canada Expression system with SAR element from IFNα2

Families Citing this family (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120258496A1 (en) 2010-09-27 2012-10-11 Boehringer Ingelheim International Gmbh Production of low fucose antibodies in h4-ii-e rat cells
HUE037811T2 (hu) 2011-10-28 2018-09-28 Prothena Biosciences Ltd Alfa-szinukleint felismerõ humanizált antitestek
EP2807188B1 (en) 2012-01-27 2019-09-11 Prothena Biosciences Limited Humanized antibodies that recognize alpha-synuclein
UA118441C2 (uk) 2012-10-08 2019-01-25 Протена Біосаєнсиз Лімітед Антитіло, що розпізнає альфа-синуклеїн
WO2014165271A2 (en) 2013-03-13 2014-10-09 Neotope Biosciences Limited Tau immunotherapy
US10513555B2 (en) 2013-07-04 2019-12-24 Prothena Biosciences Limited Antibody formulations and methods
US9951131B2 (en) 2013-07-12 2018-04-24 Prothena Biosciences Limited Antibodies that recognize IAPP
WO2015004632A1 (en) 2013-07-12 2015-01-15 Neotope Biosciences Limited Antibodies that recognize iapp
JP2017501848A (ja) 2013-11-19 2017-01-19 プロセナ バイオサイエンシーズ リミテッド 便秘症状からのレビー小体病の免疫療法のモニター
CA2938933A1 (en) 2014-03-12 2015-09-17 Prothena Biosciences Limited Anti-laminin4 antibodies specific for lg4-5
TW201623331A (zh) 2014-03-12 2016-07-01 普羅帝納生物科學公司 抗黑色素瘤細胞黏著分子(mcam)抗體類及使用彼等之相關方法
WO2015136471A1 (en) 2014-03-12 2015-09-17 Prothena Biosciences Limited Anti-laminin4 antibodies specific for lg1-3
JP6728053B2 (ja) 2014-03-12 2020-07-22 プロセナ バイオサイエンシーズ リミテッド 抗mcam抗体及び関連する使用方法
JP6744856B2 (ja) 2014-04-08 2020-08-19 プロセナ・バイオサイエンシズ・リミテッド α−シヌクレインを認識する抗体を含む血液脳関門シャトル
TWI718122B (zh) 2015-01-28 2021-02-11 愛爾蘭商普羅佘納生物科技有限公司 抗甲狀腺素運送蛋白抗體
TWI781507B (zh) 2015-01-28 2022-10-21 愛爾蘭商普羅佘納生物科技有限公司 抗甲狀腺素運送蛋白抗體
TWI711631B (zh) 2015-01-28 2020-12-01 愛爾蘭商普羅佘納生物科技有限公司 抗甲狀腺素運送蛋白抗體
RS61924B1 (sr) * 2015-02-02 2021-06-30 Meiragtx Uk Ii Ltd Regulacija genske ekspresije putem aptamerom posredovane modulacije alternativnog splajsovanja
WO2017046774A2 (en) 2015-09-16 2017-03-23 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of giant cell arteritis, polymyalgia rheumatica or takayasu's arteritis
WO2017046776A2 (en) 2015-09-16 2017-03-23 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of giant cell arteritis, polymyalgia rheumatica or takayasu's arteritis
WO2017149513A1 (en) 2016-03-03 2017-09-08 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
WO2017153955A1 (en) 2016-03-09 2017-09-14 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of granulomatous lung diseases
WO2017153953A1 (en) 2016-03-09 2017-09-14 Prothena Biosciences Limited Use of anti-mcam antibodies for treatment or prophylaxis of granulomatous lung diseases
MY197413A (en) 2016-05-02 2023-06-16 Prothena Biosciences Ltd Antibodies recognizing tau
JP7170316B2 (ja) 2016-05-02 2022-11-14 プロセナ バイオサイエンシーズ リミテッド タウ免疫療法
WO2017191560A1 (en) 2016-05-02 2017-11-09 Prothena Biosciences Limited Antibodies recognizing tau
WO2017208210A1 (en) 2016-06-03 2017-12-07 Prothena Biosciences Limited Anti-mcam antibodies and associated methods of use
JP7076711B2 (ja) 2016-07-02 2022-05-30 プロセナ バイオサイエンシーズ リミテッド 抗トランスサイレチン抗体
EP3478715A2 (en) 2016-07-02 2019-05-08 Prothena Biosciences Limited Anti-transthyretin antibodies
JP7017013B2 (ja) 2016-07-02 2022-02-08 プロセナ バイオサイエンシーズ リミテッド 抗トランスサイレチン抗体
WO2018204546A2 (en) 2017-05-02 2018-11-08 Prothena Biosciences Limited Antibodies recognizing tau
CA3076313A1 (en) 2017-09-28 2019-04-04 Prothena Biosciences Limited Dosing regimes for treatment of synucleinopathies
WO2020034097A1 (en) * 2018-08-14 2020-02-20 Wuxi Biologics (Shanghai) Co., Ltd. Transcriptional regulatory element and its use in enhancing the expression of exogenous protein
CA3117816A1 (en) 2018-11-26 2020-06-04 Forty Seven, Inc. Humanized antibodies against c-kit
BR112021016947A2 (pt) 2019-03-03 2021-11-03 Prothena Biosciences Ltd Anticorpos que reconhecem tau
JP7412860B2 (ja) 2020-10-16 2024-01-15 株式会社奥村組 泥水式シールド掘進機および泥水式シールド掘進機における圧縮空気の圧力調整方法
AU2022261882A1 (en) * 2021-04-21 2023-12-07 Janssen Biotech, Inc. Materials and methods for improved phosphotransferases

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7384744B2 (en) * 2002-11-29 2008-06-10 Boehringer Ingelheim Pharma Gmbh & Co., Kg Expression vector, methods for the production of heterologous gene products and for the selection of recombinant cells producing high levels of such products

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0173552B1 (en) 1984-08-24 1991-10-09 The Upjohn Company Recombinant dna compounds and the expression of polypeptides such as tpa
ATE289350T1 (de) 1989-04-21 2005-03-15 Amgen Inc Tnf-rezeptor, tnf bindende proteine und dafür kodierende dnas
DE69128037T2 (de) 1990-11-13 1998-05-07 Immunex Corp Bifunktionelle wählbare fusionsgene
US5786166A (en) 1991-10-25 1998-07-28 University Of Tennessee Research Corporation Methods for determining effects of a compound on the activity of bacterial periplasmic oxidoreductase enzymes
DE4228458A1 (de) 1992-08-27 1994-06-01 Beiersdorf Ag Multicistronische Expressionseinheiten und deren Verwendung
AU6953394A (en) 1993-05-21 1994-12-20 Targeted Genetics Corporation Bifunctional selectable fusion genes based on the cytosine deaminase (cd) gene
DE19539493A1 (de) 1995-10-24 1997-04-30 Thomae Gmbh Dr K Starker homologer Promotor aus Hamster
DK0873405T3 (da) * 1996-01-11 2005-01-17 Immunex Corp Ekspressionsforøgende sekvenselementer (EASE) til eukaryote ekspressionssystemer
WO2000034319A1 (en) 1998-12-11 2000-06-15 Clontech Laboratories, Inc. Fluorescent proteins from non-bioluminescent species of class anthozoa, genes encoding such proteins and uses thereof
WO2000034326A1 (en) 1998-12-11 2000-06-15 Clontech Laboratories, Inc. Fluorescent proteins from non-bioluminescent species of class anthozoa, genes encoding such proteins and uses thereof
WO2000034325A1 (en) 1998-12-11 2000-06-15 Clontech Laboratories, Inc. Fluorescent proteins from non-bioluminescent species of class anthozoa, genes encoding such proteins and uses thereof
AUPP967999A0 (en) * 1999-04-09 1999-05-06 North Western Health Care Network Viral variants
ES2257303T3 (es) 1999-07-12 2006-08-01 Genentech, Inc. Vectores de expresion y procedimientos.
EP1305412A2 (en) 1999-10-14 2003-05-02 Clontech Laboratories Inc. Anthozoa derived chromo/fluoroproteins and methods for using the same
DE60235616D1 (de) 2001-04-05 2010-04-22 Millipore Corp Gesteigerte genexpression
PT1600510E (pt) 2001-07-04 2007-06-25 Chromagenics Bv Sequências de adn com actividade anti-repressora.
US7344886B2 (en) * 2002-11-29 2008-03-18 Boehringer Ingelheim Pharma Gmbh & Co., Kg Neomycin-phosphotransferase-genes and methods for the selection of recombinant cells producing high levels of a desired gene product
DK1567652T3 (da) 2002-11-29 2007-11-26 Boehringer Ingelheim Pharma Nye neomycin-phosphotransferasegener og fremgangsmåde til selektion af höjtproducerende rekombinante celler
DE10256083A1 (de) * 2002-11-29 2004-08-12 Boehringer Ingelheim Pharma Gmbh & Co. Kg Expressionsvektor, Verfahren zur Herstellung von heterologen Genprodukten und Selektionsverfahren für hochproduzierende rekombinante Zellen

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7384744B2 (en) * 2002-11-29 2008-06-10 Boehringer Ingelheim Pharma Gmbh & Co., Kg Expression vector, methods for the production of heterologous gene products and for the selection of recombinant cells producing high levels of such products

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9085627B2 (en) 2011-04-13 2015-07-21 National Research Council Of Canada Expression system with SAR element from IFNα2

Also Published As

Publication number Publication date
AR061472A1 (es) 2008-08-27
HK1134107A1 (en) 2010-04-16
AU2007278368A1 (en) 2008-01-31
ES2401654T3 (es) 2013-04-23
JP5270544B2 (ja) 2013-08-21
CN101517085B (zh) 2013-08-07
SI2049671T1 (sl) 2013-04-30
PT2049671E (pt) 2013-01-24
US9045776B2 (en) 2015-06-02
EA200900212A1 (ru) 2009-10-30
US20150337333A1 (en) 2015-11-26
JP2009544293A (ja) 2009-12-17
EP2049671B1 (de) 2012-12-19
KR101485853B1 (ko) 2015-01-23
IL196676A0 (en) 2011-08-01
TW200815591A (en) 2008-04-01
CA2658595C (en) 2016-05-31
EP2049671A1 (de) 2009-04-22
KR20090046887A (ko) 2009-05-11
CN101517085A (zh) 2009-08-26
WO2008012142A1 (de) 2008-01-31
EA016880B1 (ru) 2012-08-30
CA2658595A1 (en) 2008-01-31
US20100311121A1 (en) 2010-12-09
BRPI0714594A2 (pt) 2013-02-19
US9708626B2 (en) 2017-07-18
AU2007278368B2 (en) 2013-12-19
CY1113711T1 (el) 2016-06-22
NO20090057L (no) 2009-02-16
PL2049671T3 (pl) 2013-05-31
MX2009000781A (es) 2009-01-29
NZ575115A (en) 2011-10-28
DK2049671T3 (da) 2013-04-08

Similar Documents

Publication Publication Date Title
US9708626B2 (en) Regulatory nucleic acid elements
AU2003302689B2 (en) Novel neomycin phosphotransferase genes and method for the selection of high-producing recombinant cells
ES2421152T3 (es) Nuevos elementos reguladores
US20090162900A1 (en) Expression vector, methods for the production of heterologous gene products and for the selection of recombinant cells producing high levels of such of such products
AU2003292102A1 (en) Expression vector, methods for the production of heterologous gene products, and selection method for high-producing recombinant cells
JP4555373B2 (ja) 新規なネオマイシンホスホトランスフェラーゼ遺伝子及び高生産組換え細胞の選抜方法
NZ540950A (en) Expression vector, methods for the production of heterologous gene products, and selection method for high-producing recombinant cells

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION

AS Assignment

Owner name: BOEHRINGER INGELHEIM PHARMA GMBH & CO. KG, GERMANY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ENENKEL, BARBARA;SAUTTER, KERSTIN;SIGNING DATES FROM 20070620 TO 20070709;REEL/FRAME:034058/0813